Skip to main content
Erschienen in: Journal of Translational Medicine 1/2024

Open Access 01.12.2024 | Review

An angel or a devil? Current view on the role of CD8+ T cells in the pathogenesis of myasthenia gravis

verfasst von: Yong Peng, Huan Yang, Quan Chen, Hong Jin, Ya-hui Xue, Miao-qiao Du, Shu Liu, Shun-yu Yao

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2024

Abstract

Background

Myasthenia gravis (MG) and the experimental autoimmune MG (EAMG) animal model are characterized by T-cell-induced and B-cell-dominated autoimmune diseases that affect the neuromuscular junction. Several subtypes of CD4+ T cells, including T helper (Th) 17 cells, follicular Th cells, and regulatory T cells (Tregs), contribute to the pathogenesis of MG. However, increasing evidence suggests that CD8+ T cells also play a critical role in the pathogenesis and treatment of MG.

Main body

Herein, we review the literature on CD8+ T cells in MG, focusing on their potential effector and regulatory roles, as well as on relevant evidence (peripheral, in situ, cerebrospinal fluid, and under different treatments), T-cell receptor usage, cytokine and chemokine expression, cell marker expression, and Treg, Tc17, CD3+CD8+CD20+ T, and CXCR5+ CD8+ T cells.

Conclusions

Further studies on CD8+ T cells in MG are necessary to determine, among others, the real pattern of the Vβ gene usage of autoantigen-specific CD8+ cells in patients with MG, real images of the physiology and function of autoantigen-specific CD8+ cells from MG/EAMG, and the subset of autoantigen-specific CD8+ cells (Tc1, Tc17, and IL-17+IFN-γ+CD8+ T cells). There are many reports of CD20-expressing T (or CD20 + T) and CXCR5+ CD8 T cells on autoimmune diseases, especially on multiple sclerosis and rheumatoid arthritis. Unfortunately, up to now, there has been no report on these T cells on MG, which might be a good direction for future studies.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Ab
Autoantibody
AChR
Muscle nicotinic acetylcholine receptor
Ag
Antigen
CSF
Cerebrospinal fluid
CTL
Cytotoxic T-lymphocytes
EAE
Experimental autoimmune encephalomyelitis
EAMG
Experimental autoimmune myasthenia gravis
EAU
Experimental autoimmune uveitis
HC
Healthy control
IFN
Interferon
IL
Interleukin
MG
Myasthenia gravis
MHC
Major histocompatibility complex
MuSK
Muscle-specific tyrosine kinase
OMG
Ocular MG
PBMCs
Peripheral blood mononuclear cells
RORγ
Retinoic acid receptor-related-orphan-receptor-C
TCR
T-cell receptors
Tfh
Follicular Th
Th
T helper
Th17
IL-17-producing CD4+ T cells
Tregs
Regulatory T cells
Tx
Thymectomy

Introduction

Myasthenia gravis (MG) and its animal model, experimental autoimmune MG (EAMG), are T-cell-driven, autoantibody (Ab)-mediated disorders affecting transmission in neuromuscular junctions [15]. As the treatment of MG has remained a burden for patients, finding better treatments is necessary [6]. Abs in patients with MG are against the muscle nicotinic acetylcholine receptor (AChR), muscle-specific tyrosine kinase (MuSK), and lipoprotein-related protein 4 [1, 3, 5, 7], and are secreted by B cells and plasma cells with assistance from CD4+ T cells [1, 8], resulting in complement anti-AChR antibodies damaging the neuromuscular junctions and inducing MG symptoms such as ptosis, dysphagia, limb weakness, and even dyspnea [4, 9].
A possible mechanism of MG might be that these autoreactive Abs bind to functional molecules in the postsynaptic membrane, impairing signal transmission in these synapses [10]. Currently, a few new Abs have been confirmed to affect patients with MG in several clinical trials. Regarding existing treatments, eculizumab has been approved in many countries [1113], zilucoplan has shown better results than eculizumab in a phase II clinical trial [14], and efgartigimod has been successful in a phase III clinical trial [15]. Moreover, immunomodulatory drugs, such as terifunomide [16, 17], have also shown benefits for MG treatment in EAMG [16].
Many subtypes of CD4+ T cells contribute to the pathogenesis of MG, including T helper 1 (Th1), Th17, Th22, follicular Th (Tfh), ThCD103, and regulatory T (Treg) cells; however, previous reports focused on CD4+ T cells [1830]. Major histocompatibility complex (MHC) class I-restricted CD8+ T cells act as both effectors and/or regulators in various autoimmune diseases, such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE) [3133], and animal models of uveitis—experimental autoimmune uveitis (EAU) [34]. However, in Theiler’s murine encephalomyelitis virus-induced demyelination, CD8+ T cells played a regulatory role predominantly. MHC class I and CD8+ T cell deficiency in β2-m−/− mice inhibited clinical scores in an animal model of type I diabetes and Ab-mediated autoimmune disease, such as systemic lupus erythematous disease.
Indeed, increasing evidence supports that CD8+ T cells also play a critical role in the pathogenesis and treatment of MG [13, 35, 36] and EAMG [37], even though this evidence is fragmented and unsystematic [38]. Herein, we investigate CD8+ T cells in MG, focusing on their potential effector and regulatory roles, as well as on evidence (peripheral, in situ, cerebrospinal fluid (CSF), and under different treatments), T-cell receptors (TCR) usage, cytokine and chemokine expression, cell marker expression, regulatory T cells, and Tc17 cells.

Evidence of CD8+ T cells in patients with MG

Peripheral CD8+ T cells in patients with MG with and without treatment

The first report on peripheral CD8+ T cells in patients with MG was published in 1988, although there were no reported significant differences in peripheral blood lymphocyte subsets (including CD4+, CD8+, and the ration of CD4+/CD8+) in patients with or without a thymus [39].
Activated CD8+ and CD4+ T cells co-exist in the peripheral blood of patients with MG, and the numbers of CD8+ and CD4+ T cells are related to the severity of the disease [40]. Furthermore, in both patients with MG and healthy controls, CD8+ peripheral blood mononuclear cells (PBMCs) and CD8+ lines responded vigorously to autologous antigen (Ag) (including AchR), which can activate CD4+ cells. The CD8+ cell lines responded equally well to the different Ag- and phytohemagglutinin-activated CD4+ cells [41]. Moreover, one report showed that the amount or function of CD8+ T cells was destroyed in patients with MG. The authors also confirmed that the inhibitory function of CD8+ cells and exogenous interleukin (IL)-2 completely restored this suppression in vitro. The expression of CD8+ was decreased in 71% of patients with MG [42]. However, another report showed that the percentages of CD8+ T cells in peripheral blood were not significantly different among three groups: ocular MG (OMG), generalized MG, and healthy controls [43].
MG usually coexists with several diseases. In autoimmune thyroid diseases, compared with thyroid antibody (TAb)-negative patients, TAb-positive patients appeared to have a higher prevalence of OMG, higher percentages of CD8+ CD28+ cells, lower AChR-Ab titers, and percentages and absolute counts of total CD8+ T cells [44]. In patients with Sjogren’s syndrome, CD8+ cells and their two subsets significantly increased in untreated patients with MG. In a female patient with MG and Good’s syndrome that presented with paroxysmal nocturnal hemoglobinuria, CD8+ T cells are directly responsible for inhibiting the growth of control PBMC. CD8+ T cells from patients with MG showed a reduced number of Vβ-TCR families (Vβ2, Vβ5.3, Vβ14, and Vβ20). CD8+ T cells are associated with damaging hematopoietic precursors as confirmed by co-culture experiments and spectratyping analyses [45].
In patients with MG, inflammatory cell infiltration was observed in the heart and skeletal muscles; however, the severity varied. The heart showed widespread inflammatory infiltrates containing multinucleated giant cells with massive myocardial degeneration. CD8+ cells were observed in both necrotic and non-necrotic muscle fibers. A limited number of mononuclear cells infiltrating the perimysial or perivascular regions were CD8+ cells. Muscle fibers, including normal-appearing ones, aberrantly express diffuse MHC class I on surface membranes (Table 1) [46].
Table 1
peripheral CD8 + T cells in patients of MG with and without treatment
Study ID
Sample
Treatment
CD8 percentage
CD4/CD8 ratio
CD8 subset
Significant findings of CD8 in MG
Machi 1988
MG
Total
39.9 ± 8.0
1.34 ± 0.43
N/A
None
 
MG
Non-Tx
39.6 ± 7.4
1.41 ± 0.42
N/A
None
 
MG
Tx
40.2 ± 9.3
1.24 ± 0.45
N/A
None
 
HC
None
39.2 ± 5.5
1.40 ± 0.34
N/A
N/A
Kawanami 1990
MG
Total
N/A
N/A
N/A
N/A
 
MG
None
29.7 ± 3.0
1.2 ± 0.16
N/A
N/A
 
MG
Tx(− )PSL( +)
26.5 ± 2.1
1.3 ± 0.18
N/A
N/A
 
MG
Tx( +) PSL( − )
25.8 ± 3.5
1.7 ± 0.31
N/A
N/A
 
MG
Tx( +) PSL( +)
29.6 ± 1.1
0.94 ± 0.24
N/A
N/A
 
HC Female
None
26.8 ± 1.7
1.5 ± 0.1
N/A
N/A
 
HC male
None
28.4 ± 1.4
1.3 ± 0.1
N/A
N/A
Shimizu 1990
MG
Total
28.0 ± 10.5
1.8 ± 1.0
CD8 + CD11 + , CD8 + CD11−
CD8 + CD11 + , CD8 + CD11−
 
MG
Tx( +) PSL( − )
22.8 ± 4.3
2.2 ± 0.8
CD8 + CD11 + , CD8 + CD11−
CD8 + CD11 + , CD8 + CD11-
 
MG
Tx( − )PSL( +)
30.8 ± 12.6
1.5 ± 1.0
CD8 + CD11 + , CD8 + CD11−
CD8 + CD11 + , CD8 + CD11-
 
MG
Tx( +) PSL( − )
25.2 ± 6.5
2.1 ± 0.9
CD8 + CD11 + , CD8 + CD11−
CD8 + CD11 + , CD8 + CD11-
 
MG
Tx( +) PSL( +)
30.2 ± 12.1
1.7 ± 1.2
CD8 + CD11 + , CD8 + CD11−
CD8 + CD11 + , CD8 + CD11−
Fujii 1991
MG group A
Tx( +) PSL( +)
39.3 ± 9.2
1.23 ± 0.36
N/A
N/A
 
MG group B
Tx( +) PSL( − )
33.3 ± 13.6
1.89 ± 0.70
N/A
N/A
 
HC
None
39.2 ± 5.5
1.40 ± 0.34
N/A
N/A
Tanaka 2009
MG
Tx( +)
Mean 21
N/A
N/A
N/A
 
MG
None
Mean 14
N/A
N/A
N/A
There have been several reports on the cell culture of CD8+ T cells in patients with MG. Out of all T-cell lines tested, CD4+ cells were only present in seven AChR-specific T-cell lines obtained from three patients with MG. In addition, predominant CD4+CD8 (15/38) and a few CD4CD8+ cells (5/38) were observed in 38 AchR or recombinant mammalian AchR α chain peptide (X4) T-cell lines obtained from 11 healthy individuals. Similarly, out of seven human AchR β subunit-specific T-cell lines obtained from patients with MG (four lines) and healthy controls (three lines), only one CD4+ and CD8+ (ratio around 1:1) were observed.

CD8+ T cells in patients with MG under different treatments

Thymoma is a non-malignant thymus tumor characterized by many non-malignant lymphocytes usually associated with MG. The percentage of mature single-positive T lymphocytes (including CD4CD8+ cells) in thymomas with MG increased significantly compared with that without MG. In addition, compared with the proportion of CD8+ cells in thymocytes of patients with MG without thymoma, those inpatients with MG with thymoma increased significantly during the incubation period with IL-2.
Thymus responses to T-cell differentiation and maturation. Thymectomy (Tx) improved the clinical outcomes in patients with MG with or without thymomas [47, 48]. After Tx and steroid therapy, CD8+ T cells decreased immediately after Tx and returned to pre-treatment levels within three weeks. These results are similar to those of other studies [4951]. Subsequently, the percentage of CD8+CD11 cells decreased, whereas that of CD8+ CD11+ cells increased after Tx and prednisolone treatment. The absolute number of CD45RA+CD8+ CD62L+ and CD45RO+CD8+T cells decreased significantly in the blood of patients with MG who underwent Tx [49]. CD8+ T cells in patients with MG increased after Tx, whereas a decrease was observed in prealbumin and albumin levels [52]. Naïve cytotoxic T cells (CD8+CD27+CD28+) were reduced in patients who underwent thymectomy [51]. CD8+ T-cell counts were markedly higher in the conventional trans-sternal extended Tx (TS) group on postoperative days 1 and 3 but remained relatively stable in the video-assisted thoracoscopic surgery group. On postoperative day 7, CD4+ counts were similar in the TS and video-assisted thoracoscopic surgery groups, whereas CD8+ counts remained higher in the TS group [53].

CD8+ T cells in patients with MG with and without thymoma

Immunosuppressive therapy, including steroids and tacrolimus, is a common treatment for MG. The percentage of DR+ and CD8+/DR+ T cells in the blood increased and the level of CD4+ T cells decreased when patients with MG received immunosuppressive therapy (steroids alone or in combination with azathioprine). Tacrolimus (FK506) is an immunosuppressive agent similar to cyclosporin A that inhibits the action of calcineurin, a serine/threonine phosphatase, thereby suppressing IL-2 production. Tacrolimus treatment significantly attenuated T-cell receptor excision circle (TREC) levels in cultured CD4CD8+ cells, but the total cell counts did not change significantly. In addition, compared with that in patients with MG without thymoma, levels of CD8+ T cells in patients with MG with thymoma decreased significantly after tacrolimus therapy [54]. Additionally, P-glycoprotein (P-gp) actively transports glucocorticoids (GC) out of target cells, thereby reducing its efficacy. Compared with patients with MG without GC therapy, P-gp function in CD8+ T cells was higher in patients with MG with long-term GC therapy (Table 2) [50].
Table 2
peripheral CD8 + T cells in patients of EAMG with and without treatment
Study ID
Sample
Treatment
CD8 percentage
CD4/CD8 ratio
CD8 subset
Significant findings of CD8 in MG
Fujii 1988
Lewis rats
Total
39.9 ± 8.0
1.34 ± 0.43
N/A
None
 
MG
Non-Tx
39.6 ± 7.4
1.41 ± 0.42
N/A
none
 
MG
Tx
40.2 ± 9.3
1.24 ± 0.45
N/A
none
 
HC
None
39.2 ± 5.5
1.40 ± 0.34
N/A
N/A
Kawanami 1990
MG
Total
N/A
N/A
N/A
N/A
 
MG
None
29.7 ± 3.0
1.2 ± 0.16
N/A
N/A
 
MG
Tx(−)PSL( +)
26.5 ± 2.1
1.3 ± 0.18
N/A
N/A
 
MG
Tx( +) PSL(−)
25.8 ± 3.5
1.7 ± 0.31
N/A
N/A
 
MG
Tx( +) PSL( +)
29.6 ± 1.1
0.94 ± 0.24
N/A
N/A
 
HC Female
None
26.8 ± 1.7
1.5 ± 0.1
N/A
N/A
 
HC male
None
28.4 ± 1.4
1.3 ± 0.1
N/A
N/A
Shimizu 1990
MG
Total
28.0 ± 10.5
1.8 ± 1.0
CD8+CD11+, CD8+CD11
CD8+CD11+, CD8+CD11
 
MG
Tx( +) PSL(−)
22.8 ± 4.3
2.2 ± 0.8
CD8+CD11+, CD8+CD11
CD8+CD11+, CD8+CD11
 
MG
Tx(−)PSL( +)
30.8 ± 12.6
1.5 ± 1.0
CD8+CD11+, CD8+CD11
CD8+CD11+, CD8+CD11
 
MG
Tx( +) PSL(−)
25.2 ± 6.5
2.1 ± 0.9
CD8+CD11+, CD8+CD11
CD8+CD11+, CD8+CD11
 
MG
Tx( +) PSL( +)
30.2 ± 12.1
1.7 ± 1.2
CD8+CD11+, CD8+CD11
CD8+CD11+, CD8+CD11
Fujii 1991
MG group A
Tx( +) PSL( +)
39.3 ± 9.2
1.23 ± 0.36
N/A
N/A
 
MG group B
Tx( +) PSL(−)
33.3 ± 13.6
1.89 ± 0.70
N/A
N/A
 
HC
None
39.2 ± 5.5
1.40 ± 0.34
N/A
N/A
Tanaka 2009
MG
Tx( +)
Mean 21
N/A
N/A
N/A
 
MG
None
Mean 14
N/A
N/A
N/A
In summary, these studies provide strong evidence of the critical role of CD8+ T cells in the pathogenesis of patients with MG with or without treatment, including patients with MG with or without other diseases, with in situ CD8+ T cells in the heart and skeletal muscle and CD8+ T cells in the CSF, as well as those undergoing different treatments, such as Tx and immunosuppressive therapy. Although CD8+ T cells are important in the pathogenesis of MG, existing studies on CD8+ T cells are fewer than those for CD4+ T cells in relation to MG. We have previously reported that the classical T-cell culture system was favorable for CD4+ T cells rather than CD8+ T cells in EAE and EAU. Possibly, the major subset of CD8+ T cells is cytotoxic T-lymphocytes (CTL), which easily causes cell damage, even to themselves, and the expansion of CD8+ T cells in vitro requires support from CD4+ T cells and their secreted cytokines [31, 32, 34, 55]. Therefore, there is a similar condition in MG/EAMG, which is also a T cell-mediated autoimmune disease.

Evidence of CD8+ T cells in EAMG

Peripheral CD8+ T cells in patients of MG with and without treatment

The first report on peripheral CD8 + T cells in EAMG was also published in 1988, although there were no reported significant differences in peripheral blood lymphocyte subsets (including CD4 + , CD8 + , and the ration of CD4 + /CD8 +) in MG with or without a thymus. Ten acetylcholine receptor (AChR)-specific T cell clones from Lewis rats were studied. These clones had various AChR subunit and peptide specificities and proliferated in response to the antigen on the appropriate APC. All T cell clones were CD4 + CD8- and OX22-, helped anti-AChR antibody production by AChR-primed lymph node B cells, and could secrete IL-2. However, several lines of evidence suggest that IL-2 was not the lymphokine that mediated T cell help. B cells primed with native AChR and exposed in culture to very low concentrations of native AChR effectively presented the Ag to T cell lines, presumably owing to an uptake via Ag receptors; however, primed B cells were no more effective than non-specific APC at presenting a synthetic AChR peptide that is recognized by AChR-specific T cells but not by AChR-specific B cells. Increasing AChR doses to a certain level produced an antibody production response that was bell shaped and only stimulated by low AChR concentrations; higher AChR concentrations suppressed the antibody production response. The evidence suggested that AChR exerted its inhibitory effect through T cells, but not via IL-2.
An EAMG model is a valuable tool for studying the pathogenesis of MG. There is evidence in rat and mouse models to support the role of CD8+ T cells in the pathogenesis of MG. Ten AchR-specific T-cell clones from Lewis rats in an EAMG model were CD4+CD8 and OX22 − . The EAMG model was generated by peritoneally injecting thymocytes from patients with MG into severe combined immunodeficiency mice, then found that in a total of 21 T-cell lines, 60% were CD4+ and 13% were CD8 + .
These results showed that the generation of CD8+ T-cell lines and clones was more difficult than that of CD4+ T cells in MG/EAMG. However, no abovementioned reports described the reason; thus, we suggest that the method of EAMG induction or cultural condition of T-cell clone selection from patients with MG or EAMG might favor CD4+ T cells, which is similar to our findings in EAE and EAU [31, 32, 34, 55].

Major TCR V gene usage of CD8+ T cells in MG

In the thymus, human thymocytes rearrange V and J segments to create a functional heterodimeric α/β TCR, which is expressed as a disulfide-linked heterodimer on the mature T cell to be recognized as antigen peptides by self-MHC. TCR V gene usage is critical for the pathogenesis of autoimmune diseases, including MG. The autoreactive CD8+ cells from patients with MG might have specific Vα and Vβ gene usage [56].
Compared with CD8+ cells from HC, several reports showed different patterns in the Vβ gene usage of CD8+ cells from patients with MG, such as (1) Vαl2.1, and Vβ6.7, 8, 12; (2) Vβ1, 13.2, 17, and 20 [56]; (3) Vβ10, 13, and 17; (4) vβ3 and Vβ19; (5) Vα2.3, 12.1, and β2, 3, 5.1, 5.2, 5.3, 6.7, 8, 13, and 17. For different subtypes of MG, remarkable CD8+ TCR Vβ-subset expansion was found in 64% and 72% of late-onset MG or thymoma-associated MG (vs. 16% with early-onset MG) [57].
The real pattern of the Vβ gene usage of CD8+ cells from patients with MG is difficult to determine because results were obtained from different groups, which might have used different standards of testing. Moreover, due to the lack of reports in the past and the last decade, this topic must be studied further.

Cytokine and chemokine secretion of CD8+ T cells in MG/EAMG

Patients with MG

In patients with MG with MuSK, tacrolimus inhibited CD8+ T-cell proliferation and interferon (IFN)-γ and IL-2 production; however, tacrolimus inhibition was lower in CD4+ T cells [58, 59]. Nevertheless, these results had two shortcomings: [1] CD8+ T cells were not highly purified using fluorescence-activated cell sorting or magnetic separation enrichment but CD3+CD8+ were only separated from whole PBMCs using flow cytometry. [2] These PBMCs were stimulated with either αCD3 and αCD28 or phorbol 12-myristate 13-acetate and ionomycin in the presence of brefeldin A but not with autoantigens such as AchR. As we have previously performed in EAE and EAU, these T cells should be dominated by non-Ag-specific T cells, which are not able to induce diseases [31, 34, 55, 60].
Intracellular staining showed that the ratio of Th2 decreased significantly, and Th1 and Th17 were significantly increased in patients with MG [24]. IL-17-producing CD4+ T cells (Th17) increased significantly with disease severity in patients with MG [36, 59]. Similarly, CD8+ T-cell subsets producing IL-17 and IL-17/IFN-γ increased in patients with MG, whereas the increased cell levels were inhibited by the retinoic acid receptor-related-orphan-receptor-C (RORγ) inhibitor. These findings provide a rationale for the exploration of targeted Th17 therapies, including ROR-γ inhibitors, to treat patients with MG [61].

EAMG

Compared with female C57BL/6 J wild-type (wt) mice, the clinical EAMG score, AChR-specific T and B cell responses, and AChR-reactive IFN-γ and IL-4-expressing cells in lymphoid organs were inhibited in CD48, CD4−/−, and CD8−/− mice. The results suggest that both CD4+ and CD8+ T cells are essential for EAMG development. Another report showed that there was earlier development of EMG and more serum anti-AChR Abs in IL-10 transgenic C57BL/6 mice and that CD8+-depleted splenocytes secreted markedly more IL-10 and lesser IFN-γ in vitro when stimulated with AChR from AChR-immunized transgenic mice [62].

T-cell marker expression of CD8+ T cells in MG

CD45RA

CD45, the tyrosine phosphatase receptor type C (PTPRC) protein, is an essential molecule involved in thymocyte maturation. CD45RA+ usually stands for the “naive” subset of CD4+ T cells, and CD45RO+ is major for “memory” T cells. Compared with HC, CD8+ but not CD4+ subsets among CD45RA+ T cells increased in the blood of patients with thymoma. Interestingly, after thymoma resection, the CD8+ but not CD4+ subset of CD45RA+ T cells in the blood decreased [63]. Furthermore, the ratio of CD45RO+ to CD45RA+ T cells was lower in CD8+ T cells of patients with early onset MG than in those of patients with late-onset MG and thymoma [64]. CD8+CD45RA+ lymphocytes in the muscle of patients with MG may signify an underlying thymoma and should not be misdiagnosed as polymyositis (PM) because all T lymphocytes in PM cases were CD45RA [65]. Furthermore, in late-onset MG, approximately 25% of the expanded cells initially exhibited a naïve CD62L+hi/CD45RA+ recent thymic emigrant-like phenotype. These expansions were significantly associated with IgG antibodies against cytomegalovirus, IL-12, and IFN-α2. The CD8+ TCRVβ expansion was stable over five years; however, recent thymic emigrant markers declined [66]. A significantly lower number of naïve helper T cells (CD4+CD45RA+) with an increased proportion of memory helper T cells (CD4+CD45RO+) and a significantly lower number of naïve cytotoxic T cells (CD8+CD27+CD28+) were observed in patients that had undergone thymectomy. Their findings indicated the premature aging of the immune system after Tx in juvenile MG; however, the associated clinical consequences could not be verified [51].

Costimulatory molecules

CD8+ cells of patients with MG expressed lower CD28 and higher CD80 and CD86. Furthermore, there were lower percentages of CD8+CD86+ Tcells in patients with an early onset of MG (< 40 years) compared with those with a late onset of MG (> 40 years). These data indicate that the CD28/CD80–CD86 costimulatory pathway is involved in MG [67]. The percentages of CD4+ and CD8+ T lymphocytes expressing the CD28 antigen were significantly lower in patients that had undergone thymectomy than in healthy subjects [68].

Chemokine receptors

Before therapy, patients with MG showed a reduction in the frequency of chemokine receptor 3 (CXCR3)+ CD4+ T cells, especially in the thymoma group; however, CXCR3+ CD8+ T cells remained normal. Conversely, after therapy, in the hyperplasia group, the expression of chemokine receptor 1 (CCR1) in CD4+ and CD8+ cells significantly increased and then returned to control levels [69]. CD4+ CXCR5+ T cells were mainly found in patients with MG; however, CD8+ T cells were predominant in HC [70]. In patients with MG, B and dendritic cells showed significantly higher levels of glycolysis and glycolytic capacity than CD8+ T cells, CD4+ T cells, and their subsets [24].

Others

Higher expression levels of estrogen receptors were observed in CD4+ helper T cells than in CD8+ cells obtained from the PBMCs of patients with MG [71]. The IFN-γ-inducible protein 10 (CXCL10) receptor and CXCR3 expression in EAMG rats increased markedly only in CD4+ and not in CD8+ T cells or CD19+ B cells [72]. Fas expression in peripheral CD8+ T cells was higher in patients with MG with a normal thymus than in patients with MG with thymoma and controls. Moreover, Fas expression in CD8+ cells was significantly higher in patients with MG treated with corticosteroids than in controls [73].
Effector CD4+ T cells expressing CD57+, CD57+ killer cell lectin-like receptor G1+, CD57+TIGIT+, CTLA-4, EOMES, FOXP3, and PD-1 increased after eculizumab treatment. However, HLA-DR expression on CD8+ T cells decreased or remained at low levels, and a mild-to-moderate-level increase in PD-1 expression was observed in CD8+ T cells after eculizumab treatment. Furthermore, C5aR was expressed on CD4+ and CD8+ T cells at low levels before and after eculizumab treatment. CD8+, CD4+CD8+, and CD4+CD8+CXCR5+ T-cell populations decreased after eculizumab treatment. CD8+ T cells expressing CD57+TIGIT + CD27low CD28low increased after eculizumab treatment [13]. In patients with MG, the expression of PI3KCA, AKT-1, mTORC1, HIF-1α, GLUT1, HK, PFK, and PK increased significantly in Th1, Th17, and CD4+ CD25 cells, and the expression of AKT-1, HIF-1α, GLUT1, PK, and CPT1A increased significantly in CD4+ CD25+ Tregs. However, there were no significant changes in the above signaling pathways in any subset of CD8+ T cells [24].

Regulatory CD8+ T cells in MG

Tregs play a critical role in regulating immune tolerance and preventing autoimmunity, and CD4+ Foxp3+ Tregs have been well-studied [74, 75]. Recent studies on CD8+ Tregs have shown good improvements [74]. Some CD8 + T cell subsets have been defined as CD8+ Tregs from different reports in different experimental systems [74]. For example, CD8+ Foxp3+ Tregs are involved in tissue transplantation and alloantigen-induced immune responses [7681]. CD8+ CD103+ cells were generated from naïve CD8+ T cells cultured with TGF-β in vitro and showed inhibition in vivo, as well as in an aggressive tumor model, and expressed Foxp3 [8284]; CD8+ CD28 T cells were found in age-dependent accumulation and chronic antigen exposure [8587]. CD8+ CD122+ CD49d+ cells express both PD-1 and IL-10 and inhibit alloantigen-induced transplant rejection [8891]. CD8+ CD122hi Ly49+ cells were discovered in young naïve mice [92, 93], EAE models [92, 94, 95], colitis [96, 97], hepatitis [98], arthritis [99], diabetes [100, 101], viral infection [102], tumor immunity [103], atherogenesis [104], and organ transplantation [105].
CD4+CD25highFoxp3+ Tregs decreased in patients with MG; however, CD8+CD28 and CD8+CD122+ Tregs do not change significantly [106]. Tacrolimus suppressed CD4+ regulatory and helper T cells in MG, such as Tregs (CD4+CD25+FOXP3+), peripheral blood Tfh (Tfh-like cells: CD4+CXCR5+), and follicular Tregs (CD4+CXCR5+FOXP3+) [59]. The percentage of CD4+CD25+CD127 Tregs in the peripheral blood of patients with generalized MG was significantly lower than that in patients with OMG and HCs [43]. The populations of CD8+CD28 and CD8+CD122+ Tregs did not differ between patients with MG and healthy controls; patients with MG exhibited a decrease in CD4+CD25highFoxp3+ Tregs and an increase in CD19+BAFF-R+ B cells, revealing that patients with MG should display the dysfunction of T-cell balance and the activation of B-cell maturation [106]. The dual APL unregulated Foxp3 expression in CD8+CD28 cells and the secretion of TGF-β and IL-10 are independent of CD8 cells. In TAChR-immunized CD8/ knockout mice, the inhibitory effect of dual APL failed, the expression of caspase 3 and 8 was unregulated, the expression of Bcl-xL was downregulated, and CD4+CD25+ Tregs were induced by dual APL. This suggests that CD8+CD28 regulatory cells play a partial role in inhibiting EAMG via dual APL [107].

CD20-expressing T cells (or CD20+ T cells) in autoimmune diseases

Anti-CD20 mAbs is a potential therapy for both antibody-mediated and T cell-mediated autoimmune diseases [108], such as MS [109112], rheumatoid arthritis (RA) [113115], systemic lupus erythematosus [115117], antineutrophil cytoplasmic antibody-associated vasculitis [117, 118], polymyositis/dermatomyositis [119, 120], primary Sjögren's syndrome [121, 122], idiopathic autoimmune thrombocytopenia and neutropenia [123], and MG [119, 124, 125]. Two mechanisms might explain the effect of rituximab on T cells: one is the indirect effect on B cell depletion that could influence T cell function, while the other, which might be more important, is the direct depletion of CD20 + T cells [126].
Typically, CD20 is expressed by mature B cells [127, 128]. However, there are some reports of CD20 being expressed by some T cells, which are called CD20-expressing T cells (CD3+ CD20+ T cells). CD20-expressing T cells were first reported in T cell lymphoma [129], RA [130] and animal-model experimental arthritis (EA) [131], MS [126, 132136], EAE [136], and neuromyelitis optical spectrum disorder (NMOSD) [132], as well as in healthy individuals (3–5% of all CD3+ cells) [137]. Furthermore, CD3+ B cells have been reported [138].

MS and EAE

In MS patients, CD20-expressing T cells have specific characteristics [132, 134, 135, 139]: located at the thymus, bone marrow, and secondary lymphatic organs, as well as CSF, even without inflammation [132, 135]; expressed CD8-related cytotoxic program (granzyme-B, Perforin, Runx3, IRF4, CD28, CD56, CD57, CD94, CD150, CD215) [139]; enriched in CD8 + and CD45RO + memory cells and in CCR7- cells [132]; high secretion of IL-4–, IL-17–, IFN-γ, and TNF-α [132]; reduced by rituximab [126], fingolimod [132], alemtuzumab [132], dimethyl fumarate [132] [135], and ocrelizumab [134], but increased by natalizumab [132]. Another study provided more evidence of CD20-expressing T cells in MS patients. At the pretreatment stage, higher frequencies of CD20dimCD8 + T cells were related to a higher concentration of myelin basic protein in CSF, higher gadolinium-lesion counts, higher T2-weighted lesion volume, and lower normal appearing white matter and thalamus volume in MRI; this might predict outcomes of anti-CD20 treatment [135]. Depletion of CD20dimCD8 + T cells could improve outcomes of anti-CD20 treatment. Furthermore, anti-CD20 treatment was not able to reconstitute CD20dimCD8 + T cells [140].
CD20 + T cells were also found in active EAE mice, and the adoptive transfer of CD20 + T cells into EAE mice enhanced the disease without the assistance of B cells [136].

RA and EA or murine collagen-induced arthritis (CIA)

In peripheral blood T cells, frequencies of CD20-expressing T cells were 0.1–6.8% in healthy individuals and 0.4–2.6% in RA patients [130]. These CD20 + T cells co-expressed CD8 (45%) and CD4 (55%), as well as differentiation/activation associated markers (CD29, CD38, CD45RO, CD49a, CD56, CD69, CD154, CD161, and CD166), but did not express other B cell markers (CD19, CD21, CD24, or soluble IgM). These CD20 + T cells secreted IFN-γ, IL-1, IL-2, IL-4, IL-8, IL-10, IL-17, MCP-1, TGF-β, and TNF-α; increased calcium flux; and easily entered apoptosis under stimulation, compared to CD20- T cells [130].

CXCR5+ CD8+ T cells in autoimmune diseases

Recently, a novel subset of CD8 T cells, CXCR5 + CD8+ T cells, were identified [141143]. CXCR5 + CD8+ T cells have been found to infiltrate the B cell follicle in response to several diseases, including viral infection (simian immunodeficiency virus (SIV) or human immunodeficiency virus (HIV) [144170], lymphocytic choriomeningitis virus (LCMV) [149, 171, 172], hepatitis B virus (HBV) [173180], polycythemia-inducing FV [180], FluA [181, 182], HSV [183], DENV2 [184, 185], and SARS CoV-2 [186190], EBV [191]); cancer (colorectal cancer [192194], NCLC [195], hepatocellular carcinoma [175, 196], hematologic malignancies [197], pancreatic tumors [198], gastric cancer [199], breast cancer [200], thyroid cancer [201], melanoma [202], and lymphoma [203]); bacterial infection (Escherichia coli, Acinetobacter baumannii, Klebsiella pneumonia, Pseudomonas aeruginosa, and Staphylococcus aureus [204]); parasitic infection (Leishmania mexicana [205]); immunodeficiency disease (CVID [206]); autoimmune diseases [4, 131, 207210] (rheumatoid arthritis (RA) [131, 207] [211], primary Sjögren syndrome (pSS) [208] [212], and multiple sclerosis (MS) [210, 213]).
CXCR5+ CD8 T cells have special phenotypes: for example, 1) cell markers including CD11a, CD11b, CD20, CD25, CD27, CD28, CD38, CD39, CD40, CD43, CD44hi, CD45RA, CD45RO, CD57, CD62L, CD69, CD83, CD94, CD95, CD101, CD103, CXCR5,CD107, CD127, CD137, CD161, CD200, CD244, HLA-DR, 41BBL, Slamf6, and TCRVa7.2 [13, 131, 145, 146, 149, 151155, 157160, 163165, 167, 168, 171, 174, 176178, 180182, 184190, 193195, 197, 199, 201, 204, 205, 207, 211, 212, 214]; 2) cytokines, chemokines and receptors (CSF-1, IL-2, IL-4, IL-6, IL-7, IL-7R, IL-10, IL-17, IL-18Ra, IL-21, IL-21R, IL-22, IL-23, IL-27, IL-27R, IL-35, IFN-I, IFNAR1, IFN-γ, TGF-β, TNF-α, IL-4R (CD124), CCR2, CCR4, CCR5, CCR6, CCR7, CCR9, CCL5, CXCL1, CXCL5, CXCL10, CXCL12 (SDF-1α), CXCL13, CXCR3, CXCR4, CXCR5, CX3CR1, MIP-1β) [144146, 149, 151, 153, 154, 157, 159162, 165, 167, 168, 170, 172174, 176, 177, 179, 181186, 188, 190, 191, 193197, 199, 200, 205, 206, 209, 211, 212]; 3) transcription factors (Bcl-6, CDCA7, CTLA-4, CULT1, E2A, pERK1/2, granulysin, granzyme B, Helios, ICOS, Id2, ISGs, interferon pathway associated molecules (MX1, MX2, GBP1, and ISG15), Ki-67, KLRG1, LAG-3, MAMU-DRA, MEF2C, NFATC1, NFATC2, PD-1, PD-L1, PD-L2, perforin, PRDM1, RANTES, SOX4, SPRY2, STAT2, STAT6, TCF4, TCF24, and Tim-3) [145, 146, 148, 149, 151158, 162166, 168, 172, 174, 175, 178, 211].

Conclusion

In this review, we focused on the updated information on CD8+ T cells in MG/EAMG, as well as on the relevant evidence (peripheral, in situ, CSF, and under different conditions), in vitro culture, TCR usage, cytokine and chemokine expression, cell marker expression, Tregs, and Tc17. However, the mechanism underlying the regulatory role of CD8+ T cells in MG/EAMG remains unclear. In addition, the following questions should be addressed in future studies: (1) Why are fewer studies available on CD8+ T cells in MG than those on CD4+ T cells in MG?; (2) Is the classical T-cell culture system favorable to CD4+ T cells compared to CD8+ T cells, and is it similar to our experience with studies on CD8+ T cells in EAE and EAU [31, 32, 34, 55]?; (3) What is the real pattern of the Vβ gene usage of autoantigen-specific CD8+ cells from patients with MG?; (4) As CD8+ T cells were not highly purified and were not stimulated by autoantigens, what are the real images of the physiology and function of autoantigen-specific CD8+ cells from MG/EAMG available?; (5) Which subset of autoantigen-specific CD8+ cells (Tc1, Tc17, IL-17+IFN-γ+CD8+ T cells, or IL-17+IFN-γ+TNF-α+CD8+ T cells) plays the most critical role in the pathogenesis of MG/EAMG? Did they like the similar studies that have been performed on EAE, diabetes, and systemic lupus erythematous disease [32]; (6) Which subsets of autoantigen-specific CD8+ Tregs play a regulatory role in MG/EAMG among CD8+CD28, CD8+CD122+ [106], CD8+ CD122+ CD49d+ cells [8891], or CD8+ CD122hi Ly49+ cells [92105], which have been well studied in other diseases. Hence, it is necessary to perform further studies on CD8+ T cells in MG/EAMG, especially on autoantigen-specific CD8+ Tregs.
In additional, there are many reports of CD20-expressing T cells (or CD20 + T cells) and CXCR5+ CD8 T cells on autoimmune diseases, especially on MS and RA. Unfortunately, up to now, there has been no report on these T cells on MG, which may be a good direction for future studies on MG.

Acknowledgements

Not applicable.

Declarations

This is a review, hence, not applicable.
Not applicable.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Gilhus NE, Verschuuren JJ. Myasthenia gravis: subgroup classification and therapeutic strategies. Lancet Neurol. 2015;14(10):1023–36.PubMedCrossRef Gilhus NE, Verschuuren JJ. Myasthenia gravis: subgroup classification and therapeutic strategies. Lancet Neurol. 2015;14(10):1023–36.PubMedCrossRef
2.
Zurück zum Zitat Jin W, Luo Z, Yang H. Peripheral B cell subsets in autoimmune diseases: clinical implications and effects of B cell-targeted therapies. J Immunol Res. 2020;2020:9518137.PubMedPubMedCentralCrossRef Jin W, Luo Z, Yang H. Peripheral B cell subsets in autoimmune diseases: clinical implications and effects of B cell-targeted therapies. J Immunol Res. 2020;2020:9518137.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Song J, Zhao R, Yan C, Luo S, Xi J, Ding P, et al. A targeted complement inhibitor CRIg/FH protects against experimental autoimmune myasthenia gravis in rats via immune modulation. Front Immunol. 2022;13: 746068.PubMedPubMedCentralCrossRef Song J, Zhao R, Yan C, Luo S, Xi J, Ding P, et al. A targeted complement inhibitor CRIg/FH protects against experimental autoimmune myasthenia gravis in rats via immune modulation. Front Immunol. 2022;13: 746068.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Fan R, Que W, Liu Z, Zheng W, Guo X, Liu L, et al. Single-cell mapping reveals dysregulation of immune cell populations and VISTA+ monocytes in myasthenia gravis. Clin Immunol. 2022;245: 109184.PubMedCrossRef Fan R, Que W, Liu Z, Zheng W, Guo X, Liu L, et al. Single-cell mapping reveals dysregulation of immune cell populations and VISTA+ monocytes in myasthenia gravis. Clin Immunol. 2022;245: 109184.PubMedCrossRef
5.
Zurück zum Zitat Zhong H, Jiao K, Huan X, Zhao R, Su M, Goh LY, et al. Herpesvirus entry mediator on T cells as a protective factor for myasthenia gravis: a mendelian randomization study. Front Immunol. 2022;13: 931821.PubMedPubMedCentralCrossRef Zhong H, Jiao K, Huan X, Zhao R, Su M, Goh LY, et al. Herpesvirus entry mediator on T cells as a protective factor for myasthenia gravis: a mendelian randomization study. Front Immunol. 2022;13: 931821.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Cutter G, Xin H, Aban I, Burns TM, Allman PH, Farzaneh-Far R, et al. Cross-sectional analysis of the Myasthenia Gravis patient registry: disability and treatment. Muscle Nerve. 2019;60(6):707–15.PubMedPubMedCentralCrossRef Cutter G, Xin H, Aban I, Burns TM, Allman PH, Farzaneh-Far R, et al. Cross-sectional analysis of the Myasthenia Gravis patient registry: disability and treatment. Muscle Nerve. 2019;60(6):707–15.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Gilhus NE, Tzartos S, Evoli A, Palace J, Burns TM, Verschuuren J. Myasthenia gravis. Nat Rev Dis Primers. 2019;5(1):30.PubMedCrossRef Gilhus NE, Tzartos S, Evoli A, Palace J, Burns TM, Verschuuren J. Myasthenia gravis. Nat Rev Dis Primers. 2019;5(1):30.PubMedCrossRef
8.
Zurück zum Zitat Dalakas MC. Immunotherapy in myasthenia gravis in the era of biologics. Nat Rev Neurol. 2019;15(2):113–24.PubMedCrossRef Dalakas MC. Immunotherapy in myasthenia gravis in the era of biologics. Nat Rev Neurol. 2019;15(2):113–24.PubMedCrossRef
9.
Zurück zum Zitat Uzawa A, Kuwabara S, Suzuki S, Imai T, Murai H, Ozawa Y, et al. Roles of cytokines and T cells in the pathogenesis of myasthenia gravis. Clin Exp Immunol. 2021;203(3):366–74.PubMedCrossRef Uzawa A, Kuwabara S, Suzuki S, Imai T, Murai H, Ozawa Y, et al. Roles of cytokines and T cells in the pathogenesis of myasthenia gravis. Clin Exp Immunol. 2021;203(3):366–74.PubMedCrossRef
10.
Zurück zum Zitat Carr AS, Cardwell CR, McCarron PO, McConville J. A systematic review of population based epidemiological studies in Myasthenia Gravis. BMC Neurol. 2010;10:46.PubMedPubMedCentralCrossRef Carr AS, Cardwell CR, McCarron PO, McConville J. A systematic review of population based epidemiological studies in Myasthenia Gravis. BMC Neurol. 2010;10:46.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Howard JF Jr, Utsugisawa K, Benatar M, Murai H, Barohn RJ, Illa I, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol. 2017;16(12):976–86.PubMedCrossRef Howard JF Jr, Utsugisawa K, Benatar M, Murai H, Barohn RJ, Illa I, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol. 2017;16(12):976–86.PubMedCrossRef
12.
Zurück zum Zitat Mantegazza R, Wolfe GI, Muppidi S, Wiendl H, Fujita KP, O’Brien FL, et al. Post-intervention status in patients with refractory myasthenia gravis treated with eculizumab during REGAIN and Its open-label extension. Neurology. 2021;96(4):e610–8.PubMedPubMedCentralCrossRef Mantegazza R, Wolfe GI, Muppidi S, Wiendl H, Fujita KP, O’Brien FL, et al. Post-intervention status in patients with refractory myasthenia gravis treated with eculizumab during REGAIN and Its open-label extension. Neurology. 2021;96(4):e610–8.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Li Y, Yi JS, Howard JF Jr, Chopra M, Russo MA, Guptill JT. Cellular changes in eculizumab early responders with generalized myasthenia gravis. Clin Immunol. 2021;231: 108830.PubMedCrossRef Li Y, Yi JS, Howard JF Jr, Chopra M, Russo MA, Guptill JT. Cellular changes in eculizumab early responders with generalized myasthenia gravis. Clin Immunol. 2021;231: 108830.PubMedCrossRef
14.
Zurück zum Zitat Howard JF Jr, Nowak RJ, Wolfe GI, Freimer ML, Vu TH, Hinton JL, et al. Clinical effects of the self-administered subcutaneous complement inhibitor zilucoplan in patients with moderate to severe generalized myasthenia gravis: results of a phase 2 randomized, double-blind, placebo-controlled. Multicenter Clin Trial JAMA Neurol. 2020;77(5):582–92.CrossRef Howard JF Jr, Nowak RJ, Wolfe GI, Freimer ML, Vu TH, Hinton JL, et al. Clinical effects of the self-administered subcutaneous complement inhibitor zilucoplan in patients with moderate to severe generalized myasthenia gravis: results of a phase 2 randomized, double-blind, placebo-controlled. Multicenter Clin Trial JAMA Neurol. 2020;77(5):582–92.CrossRef
15.
Zurück zum Zitat Howard JF Jr, Bril V, Vu T, Karam C, Peric S, Margania T, et al. Safety, efficacy, and tolerability of efgartigimod in patients with generalised myasthenia gravis (ADAPT): a multicentre, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 2021;20(7):526–36.PubMedCrossRef Howard JF Jr, Bril V, Vu T, Karam C, Peric S, Margania T, et al. Safety, efficacy, and tolerability of efgartigimod in patients with generalised myasthenia gravis (ADAPT): a multicentre, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 2021;20(7):526–36.PubMedCrossRef
16.
Zurück zum Zitat Koseoglu E, Sungur N, Muhtaroglu S, Zararsiz G, Eken A. The beneficial clinical effects of teriflunomide in experimental autoimmune myasthenia gravis and the investigation of the possible immunological mechanisms. Cell Mol Neurobiol. 2022;43(5):2071.PubMedCrossRef Koseoglu E, Sungur N, Muhtaroglu S, Zararsiz G, Eken A. The beneficial clinical effects of teriflunomide in experimental autoimmune myasthenia gravis and the investigation of the possible immunological mechanisms. Cell Mol Neurobiol. 2022;43(5):2071.PubMedCrossRef
17.
Zurück zum Zitat Yilmaz V, Ulusoy C, Hajtovic S, Turkoglu R, Kurtuncu M, Tzartos J, et al. Effects of teriflunomide on B cell subsets in MuSK-induced experimental autoimmune myasthenia gravis and multiple sclerosis. Immunol Invest. 2021;50(6):671–84.PubMedCrossRef Yilmaz V, Ulusoy C, Hajtovic S, Turkoglu R, Kurtuncu M, Tzartos J, et al. Effects of teriflunomide on B cell subsets in MuSK-induced experimental autoimmune myasthenia gravis and multiple sclerosis. Immunol Invest. 2021;50(6):671–84.PubMedCrossRef
18.
Zurück zum Zitat Uzawa A, Kawaguchi N, Kanai T, Himuro K, Kuwabara S. Serum high mobility group box 1 is upregulated in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2015;86(6):695–7.PubMedCrossRef Uzawa A, Kawaguchi N, Kanai T, Himuro K, Kuwabara S. Serum high mobility group box 1 is upregulated in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2015;86(6):695–7.PubMedCrossRef
19.
Zurück zum Zitat Uzawa A, Kawaguchi N, Kanai T, Himuro K, Oda F, Kuwabara S. Increased serum peroxiredoxin 5 levels in myasthenia gravis. J Neuroimmunol. 2015;287:16–8.PubMedCrossRef Uzawa A, Kawaguchi N, Kanai T, Himuro K, Oda F, Kuwabara S. Increased serum peroxiredoxin 5 levels in myasthenia gravis. J Neuroimmunol. 2015;287:16–8.PubMedCrossRef
20.
Zurück zum Zitat Xie Y, Li HF, Jiang B, Li Y, Kaminski HJ, Kusner LL. Elevated plasma interleukin-17A in a subgroup of myasthenia Gravis patients. Cytokine. 2016;78:44–6.PubMedCrossRef Xie Y, Li HF, Jiang B, Li Y, Kaminski HJ, Kusner LL. Elevated plasma interleukin-17A in a subgroup of myasthenia Gravis patients. Cytokine. 2016;78:44–6.PubMedCrossRef
21.
Zurück zum Zitat Zhang CJ, Gong Y, Zhu W, Qi Y, Yang CS, Fu Y, et al. Augmentation of circulating follicular helper T cells and their impact on autoreactive B cells in myasthenia gravis. J Immunol. 2016;197(7):2610–7.PubMedCrossRef Zhang CJ, Gong Y, Zhu W, Qi Y, Yang CS, Fu Y, et al. Augmentation of circulating follicular helper T cells and their impact on autoreactive B cells in myasthenia gravis. J Immunol. 2016;197(7):2610–7.PubMedCrossRef
22.
Zurück zum Zitat Zhang X, Liu S, Chang T, Xu J, Zhang C, Tian F, et al. Intrathymic Tfh/B cells interaction leads to ectopic GCs formation and anti-AChR antibody production: central role in triggering MG occurrence. Mol Neurobiol. 2016;53(1):120–31.PubMedCrossRef Zhang X, Liu S, Chang T, Xu J, Zhang C, Tian F, et al. Intrathymic Tfh/B cells interaction leads to ectopic GCs formation and anti-AChR antibody production: central role in triggering MG occurrence. Mol Neurobiol. 2016;53(1):120–31.PubMedCrossRef
23.
Zurück zum Zitat Kohler S, Keil TOP, Hoffmann S, Swierzy M, Ismail M, Rückert JC, et al. CD4(+) FoxP3(+) T regulatory cell subsets in myasthenia gravis patients. Clin Immunol. 2017;179:40–6.PubMedCrossRef Kohler S, Keil TOP, Hoffmann S, Swierzy M, Ismail M, Rückert JC, et al. CD4(+) FoxP3(+) T regulatory cell subsets in myasthenia gravis patients. Clin Immunol. 2017;179:40–6.PubMedCrossRef
24.
Zurück zum Zitat Li Z, Peng Y, Li Y, Zhou R, Chen D, Jin W, et al. Glucose metabolism pattern of peripheral blood immune cells in myasthenia gravis patients. Ann Transl Med. 2020;8(9):577.PubMedPubMedCentralCrossRef Li Z, Peng Y, Li Y, Zhou R, Chen D, Jin W, et al. Glucose metabolism pattern of peripheral blood immune cells in myasthenia gravis patients. Ann Transl Med. 2020;8(9):577.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Li Y, Yang S, Li Z, Meng H, Jin W, Yang H, et al. Soluble glucocorticoid-induced tumor necrosis factor receptor regulates Helios expression in myasthenia gravis. J Transl Med. 2019;17(1):168.PubMedPubMedCentralCrossRef Li Y, Yang S, Li Z, Meng H, Jin W, Yang H, et al. Soluble glucocorticoid-induced tumor necrosis factor receptor regulates Helios expression in myasthenia gravis. J Transl Med. 2019;17(1):168.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Wen Y, Yang B, Lu J, Zhang J, Yang H, Li J. Imbalance of circulating CD4(+)CXCR5(+)FOXP3(+) Tfr-like cells and CD4(+)CXCR5(+)FOXP3(-) Tfh-like cells in myasthenia gravis. Neurosci Lett. 2016;630:176–82.PubMedCrossRef Wen Y, Yang B, Lu J, Zhang J, Yang H, Li J. Imbalance of circulating CD4(+)CXCR5(+)FOXP3(+) Tfr-like cells and CD4(+)CXCR5(+)FOXP3(-) Tfh-like cells in myasthenia gravis. Neurosci Lett. 2016;630:176–82.PubMedCrossRef
27.
Zurück zum Zitat Zhang J, Chen Y, Jia G, Chen X, Lu J, Yang H, et al. FOXP3 -3279 and IVS9+459 polymorphisms are associated with genetic susceptibility to myasthenia gravis. Neurosci Lett. 2013;534:274–8.PubMedCrossRef Zhang J, Chen Y, Jia G, Chen X, Lu J, Yang H, et al. FOXP3 -3279 and IVS9+459 polymorphisms are associated with genetic susceptibility to myasthenia gravis. Neurosci Lett. 2013;534:274–8.PubMedCrossRef
28.
Zurück zum Zitat Yang G, Yang X, Zhang J, Li G, Zheng D, Peng A, et al. Transcriptional repressor Blimp1 regulates follicular regulatory T-cell homeostasis and function. Immunology. 2018;153(1):105–17.PubMedCrossRef Yang G, Yang X, Zhang J, Li G, Zheng D, Peng A, et al. Transcriptional repressor Blimp1 regulates follicular regulatory T-cell homeostasis and function. Immunology. 2018;153(1):105–17.PubMedCrossRef
29.
Zurück zum Zitat Huan X, Luo S, Zhong H, Zheng X, Song J, Zhou L, et al. In-depth peripheral CD4(+) T profile correlates with myasthenic crisis. Ann Clin Transl Neurol. 2021;8(4):749–62.PubMedPubMedCentralCrossRef Huan X, Luo S, Zhong H, Zheng X, Song J, Zhou L, et al. In-depth peripheral CD4(+) T profile correlates with myasthenic crisis. Ann Clin Transl Neurol. 2021;8(4):749–62.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Peng Y, Zhu FZ, Chen ZX, Zhou JX, Gan L, Yang SS, et al. Characterization of myelin oligodendrocyte glycoprotein (MOG)35–55-specific CD8+ T cells in experimental autoimmune encephalomyelitis. Chin Med J. 2019;132(24):2934–40.PubMedPubMedCentralCrossRef Peng Y, Zhu FZ, Chen ZX, Zhou JX, Gan L, Yang SS, et al. Characterization of myelin oligodendrocyte glycoprotein (MOG)35–55-specific CD8+ T cells in experimental autoimmune encephalomyelitis. Chin Med J. 2019;132(24):2934–40.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Peng YZF, Deng X, Zhou JX, Gao S, Chen ZX, et al. Experimental autoimmune encephalomyelitis inhibited by Huangqi guizhi wuwu decoction via Th2 cytokine enhancement. World J Tradit Chin Med. 2021;7:467–76.CrossRef Peng YZF, Deng X, Zhou JX, Gao S, Chen ZX, et al. Experimental autoimmune encephalomyelitis inhibited by Huangqi guizhi wuwu decoction via Th2 cytokine enhancement. World J Tradit Chin Med. 2021;7:467–76.CrossRef
34.
Zurück zum Zitat Peng Y, Shao H, Ke Y, Zhang P, Han G, Kaplan HJ. Minimally activated CD8 autoreactive T cells specific for IRBP express a high level of Foxp3 and are functionally suppressive. Invest Ophthalmol Vis Sci. 2007;48(5):2178–84.PubMedCrossRef Peng Y, Shao H, Ke Y, Zhang P, Han G, Kaplan HJ. Minimally activated CD8 autoreactive T cells specific for IRBP express a high level of Foxp3 and are functionally suppressive. Invest Ophthalmol Vis Sci. 2007;48(5):2178–84.PubMedCrossRef
35.
Zurück zum Zitat Robat-Jazi B, Hosseini M, Shaygannejad V, Nafissi S, Rezaei A, Mansourain M, et al. High frequency of Tc22 and Th22 cells in myasthenia gravis patients and their significant reduction after thymectomy. NeuroImmunoModulation. 2018;25(2):80–8.PubMedCrossRef Robat-Jazi B, Hosseini M, Shaygannejad V, Nafissi S, Rezaei A, Mansourain M, et al. High frequency of Tc22 and Th22 cells in myasthenia gravis patients and their significant reduction after thymectomy. NeuroImmunoModulation. 2018;25(2):80–8.PubMedCrossRef
36.
Zurück zum Zitat Hosseini M, Robat-Jazi B, Shaygannejad V, Naffisi S, Mirmossayeb O, Rezaei A, et al. Increased proportion of Tc17 and Th17 cells and their significant reduction after thymectomy may be related to disease progression in Myasthenia Gravis. NeuroImmuno Modulation. 2017;24(4–5):264–70.CrossRef Hosseini M, Robat-Jazi B, Shaygannejad V, Naffisi S, Mirmossayeb O, Rezaei A, et al. Increased proportion of Tc17 and Th17 cells and their significant reduction after thymectomy may be related to disease progression in Myasthenia Gravis. NeuroImmuno Modulation. 2017;24(4–5):264–70.CrossRef
37.
Zurück zum Zitat Qin HBLH, Jia CY, Zhang XF, Zhang XL, Cheng M. The regulation of interleukin-7/CD127 signaling pathway on CD8+ T cells in patients with myasthenia gravis. Chin J Neurol. 2022;55(6):597–604. Qin HBLH, Jia CY, Zhang XF, Zhang XL, Cheng M. The regulation of interleukin-7/CD127 signaling pathway on CD8+ T cells in patients with myasthenia gravis. Chin J Neurol. 2022;55(6):597–604.
38.
Zurück zum Zitat Zhong H, Zhao C, Luo S. HLA in myasthenia gravis: from superficial correlation to underlying mechanism. Autoimmun Rev. 2019;18(9): 102349.PubMedCrossRef Zhong H, Zhao C, Luo S. HLA in myasthenia gravis: from superficial correlation to underlying mechanism. Autoimmun Rev. 2019;18(9): 102349.PubMedCrossRef
39.
Zurück zum Zitat Machi M, Itoyama Y, Goto I, Kuroiwa Y. Surface phenotypes of lymphoid cells altered in the human myasthenic thymus. Neurology. 1988;38(4):592–6.PubMedCrossRef Machi M, Itoyama Y, Goto I, Kuroiwa Y. Surface phenotypes of lymphoid cells altered in the human myasthenic thymus. Neurology. 1988;38(4):592–6.PubMedCrossRef
40.
Zurück zum Zitat Protti MP, Manfredi AA, Straub C, Howard JF Jr, Conti-Tronconi BM. CD4+ T cell response to the human acetylcholine receptor alpha subunit in myasthenia gravis a study with synthetic peptides. J Immunol. 1990;144(4):1276–81.PubMedCrossRef Protti MP, Manfredi AA, Straub C, Howard JF Jr, Conti-Tronconi BM. CD4+ T cell response to the human acetylcholine receptor alpha subunit in myasthenia gravis a study with synthetic peptides. J Immunol. 1990;144(4):1276–81.PubMedCrossRef
41.
Zurück zum Zitat Protti MP, Manfredi AA, Wu XD, Moiola L, Howard JF Jr, Conti-Tronconi BM. Myasthenia gravis T epitopes on the delta subunit of human muscle acetylcholine receptor. J Immunol. 1991;146(7):2253–61.PubMedCrossRef Protti MP, Manfredi AA, Wu XD, Moiola L, Howard JF Jr, Conti-Tronconi BM. Myasthenia gravis T epitopes on the delta subunit of human muscle acetylcholine receptor. J Immunol. 1991;146(7):2253–61.PubMedCrossRef
42.
43.
Zurück zum Zitat Hu Y, Wang J, Rao J, Xu X, Cheng Y, Yan L, et al. Comparison of peripheral blood B cell subset ratios and B cell-related cytokine levels between ocular and generalized myasthenia gravis. Int Immunopharmacol. 2020;80: 106130.PubMedCrossRef Hu Y, Wang J, Rao J, Xu X, Cheng Y, Yan L, et al. Comparison of peripheral blood B cell subset ratios and B cell-related cytokine levels between ocular and generalized myasthenia gravis. Int Immunopharmacol. 2020;80: 106130.PubMedCrossRef
44.
Zurück zum Zitat Zhang Q, Bi Z, Yang M, Gui M, Bu B. Differences in immunophenotypes between myasthenia gravis patients with and without thyroid antibodies. Muscle Nerve. 2022;65(5):553–9.PubMedCrossRef Zhang Q, Bi Z, Yang M, Gui M, Bu B. Differences in immunophenotypes between myasthenia gravis patients with and without thyroid antibodies. Muscle Nerve. 2022;65(5):553–9.PubMedCrossRef
45.
Zurück zum Zitat Palmieri G, Selleri C, Montella L, Bulgarelli G, Vitiello L, Merkabaoui G, et al. Thymoma followed by paroxysmal nocturnal hemoglobinuria: a unique clinical association in the context of multiorgan autoimmunity with a potential role for CD8+ T lymphocytes. Am J Hematol. 2006;81(10):774–8.PubMedCrossRef Palmieri G, Selleri C, Montella L, Bulgarelli G, Vitiello L, Merkabaoui G, et al. Thymoma followed by paroxysmal nocturnal hemoglobinuria: a unique clinical association in the context of multiorgan autoimmunity with a potential role for CD8+ T lymphocytes. Am J Hematol. 2006;81(10):774–8.PubMedCrossRef
46.
Zurück zum Zitat Suzuki S, Utsugisawa K, Yoshikawa H, Motomura M, Matsubara S, Yokoyama K, et al. Autoimmune targets of heart and skeletal muscles in myasthenia gravis. Arch Neurol. 2009;66(11):1334–8.PubMedCrossRef Suzuki S, Utsugisawa K, Yoshikawa H, Motomura M, Matsubara S, Yokoyama K, et al. Autoimmune targets of heart and skeletal muscles in myasthenia gravis. Arch Neurol. 2009;66(11):1334–8.PubMedCrossRef
47.
Zurück zum Zitat Wolfe GI, Kaminski HJ, Aban IB, Minisman G, Kuo HC, Marx A, et al. Randomized trial of thymectomy in Myasthenia Gravis. N Engl J Med. 2016;375(6):511–22.PubMedPubMedCentralCrossRef Wolfe GI, Kaminski HJ, Aban IB, Minisman G, Kuo HC, Marx A, et al. Randomized trial of thymectomy in Myasthenia Gravis. N Engl J Med. 2016;375(6):511–22.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Wolfe GI, Kaminski HJ, Aban IB, Minisman G, Kuo HC, Marx A, et al. Long-term effect of thymectomy plus prednisone versus prednisone alone in patients with non-thymomatous myasthenia gravis: 2-year extension of the MGTX randomised trial. Lancet Neurol. 2019;18(3):259–68.PubMedPubMedCentralCrossRef Wolfe GI, Kaminski HJ, Aban IB, Minisman G, Kuo HC, Marx A, et al. Long-term effect of thymectomy plus prednisone versus prednisone alone in patients with non-thymomatous myasthenia gravis: 2-year extension of the MGTX randomised trial. Lancet Neurol. 2019;18(3):259–68.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Sempowski G, Thomasch J, Gooding M, Hale L, Edwards L, Ciafaloni E, et al. Effect of thymectomy on human peripheral blood T cell pools in myasthenia gravis. J Immunol. 2001;166(4):2808–17.PubMedCrossRef Sempowski G, Thomasch J, Gooding M, Hale L, Edwards L, Ciafaloni E, et al. Effect of thymectomy on human peripheral blood T cell pools in myasthenia gravis. J Immunol. 2001;166(4):2808–17.PubMedCrossRef
50.
Zurück zum Zitat Tanaka S, Masuda M, Nakajima K, Ido N, Ohtsuka T, Nishida M, et al. P-glycoprotein function in peripheral T lymphocyte subsets of myasthenia gravis patients: clinical implications and influence of glucocorticoid administration. Int Immunopharmacol. 2009;9(3):284–90.PubMedCrossRef Tanaka S, Masuda M, Nakajima K, Ido N, Ohtsuka T, Nishida M, et al. P-glycoprotein function in peripheral T lymphocyte subsets of myasthenia gravis patients: clinical implications and influence of glucocorticoid administration. Int Immunopharmacol. 2009;9(3):284–90.PubMedCrossRef
51.
Zurück zum Zitat Popperud TH, Gul KA, Brunborg C, Olaussen RW, Abrahamsen TG, Osnes LT, et al. Thymectomy in juvenile myasthenia gravis is safe regarding long term immunological effects. Front Neurol. 2021;12: 596859.PubMedPubMedCentralCrossRef Popperud TH, Gul KA, Brunborg C, Olaussen RW, Abrahamsen TG, Osnes LT, et al. Thymectomy in juvenile myasthenia gravis is safe regarding long term immunological effects. Front Neurol. 2021;12: 596859.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Xin Y, Cai H, Wu L, Cui Y. The Effect of immunonutrition on the postoperative complications in thymoma with myasthenia gravis. Mediators Inflamm. 2016;2016:8781740.PubMedPubMedCentralCrossRef Xin Y, Cai H, Wu L, Cui Y. The Effect of immunonutrition on the postoperative complications in thymoma with myasthenia gravis. Mediators Inflamm. 2016;2016:8781740.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Chen B, Wang Y, Geng Y, Huang Y, Guo S, Mao X. Marked improvement of anti-N-methyl-D-aspartate receptor encephalitis by large-dose methylprednisolone and plasmapheresis therapy combined with (18)F-fluorodeoxyglucose positron emission tomography imaging: a case report. Exp Ther Med. 2014;8(4):1167–9.PubMedPubMedCentralCrossRef Chen B, Wang Y, Geng Y, Huang Y, Guo S, Mao X. Marked improvement of anti-N-methyl-D-aspartate receptor encephalitis by large-dose methylprednisolone and plasmapheresis therapy combined with (18)F-fluorodeoxyglucose positron emission tomography imaging: a case report. Exp Ther Med. 2014;8(4):1167–9.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Mitsui T, Kuroda Y, Ueno S, Matsui N, Kaji R. FK506 attenuates thymic output in patients with myasthenia gravis. Arch Med Sci. 2013;9(6):1090–6.PubMedPubMedCentralCrossRef Mitsui T, Kuroda Y, Ueno S, Matsui N, Kaji R. FK506 attenuates thymic output in patients with myasthenia gravis. Arch Med Sci. 2013;9(6):1090–6.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Peng Y, Zhu F-Z, Deng X, Zhou J-X, Gao S, Chen Z-X, et al. Experimental autoimmune encephalomyelitis inhibited by huangqi guizhi wuwu decoction via th2 cytokine enhancement. World J Tradit Chin Med. 2021;7(4):467–76.CrossRef Peng Y, Zhu F-Z, Deng X, Zhou J-X, Gao S, Chen Z-X, et al. Experimental autoimmune encephalomyelitis inhibited by huangqi guizhi wuwu decoction via th2 cytokine enhancement. World J Tradit Chin Med. 2021;7(4):467–76.CrossRef
56.
Zurück zum Zitat Infante AJ, Baillargeon J, Kraig E, Lott L, Jackson C, Hämmerling GJ, et al. Evidence of a diverse T cell receptor repertoire for acetylcholine receptor, the autoantigen of myasthenia gravis. J Autoimmun. 2003;21(2):167–74.PubMedCrossRef Infante AJ, Baillargeon J, Kraig E, Lott L, Jackson C, Hämmerling GJ, et al. Evidence of a diverse T cell receptor repertoire for acetylcholine receptor, the autoantigen of myasthenia gravis. J Autoimmun. 2003;21(2):167–74.PubMedCrossRef
57.
Zurück zum Zitat Tackenberg B, Kruth J, Bartholomaeus JE, Schlegel K, Oertel WH, Willcox N, et al. Clonal expansions of CD4+ B helper T cells in autoimmune myasthenia gravis. Eur J Immunol. 2007;37(3):849–63.PubMedCrossRef Tackenberg B, Kruth J, Bartholomaeus JE, Schlegel K, Oertel WH, Willcox N, et al. Clonal expansions of CD4+ B helper T cells in autoimmune myasthenia gravis. Eur J Immunol. 2007;37(3):849–63.PubMedCrossRef
58.
Zurück zum Zitat Yi JS, Guidon A, Sparks S, Osborne R, Juel VC, Massey JM, et al. Characterization of CD4 and CD8 T cell responses in MuSK myasthenia gravis. J Autoimmun. 2014;52:130–8.PubMedCrossRef Yi JS, Guidon A, Sparks S, Osborne R, Juel VC, Massey JM, et al. Characterization of CD4 and CD8 T cell responses in MuSK myasthenia gravis. J Autoimmun. 2014;52:130–8.PubMedCrossRef
59.
Zurück zum Zitat Li Y, Guptill JT, Russo MA, Massey JM, Juel VC, Hobson-Webb LD, et al. Tacrolimus inhibits Th1 and Th17 responses in MuSK-antibody positive myasthenia gravis patients. Exp Neurol. 2019;312:43–50.PubMedCrossRef Li Y, Guptill JT, Russo MA, Massey JM, Juel VC, Hobson-Webb LD, et al. Tacrolimus inhibits Th1 and Th17 responses in MuSK-antibody positive myasthenia gravis patients. Exp Neurol. 2019;312:43–50.PubMedCrossRef
60.
Zurück zum Zitat Peng Y, Han G, Shao H, Wang Y, Kaplan HJ, Sun D. Characterization of IL-17(+) interphotoreceptor retinoid-binding protein-specific T cells in experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2007;48(9):4153–61.PubMedCrossRef Peng Y, Han G, Shao H, Wang Y, Kaplan HJ, Sun D. Characterization of IL-17(+) interphotoreceptor retinoid-binding protein-specific T cells in experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2007;48(9):4153–61.PubMedCrossRef
61.
Zurück zum Zitat Yi JS, Russo MA, Raja S, Massey JM, Juel VC, Shin J, et al. Inhibition of the transcription factor ROR-γ reduces pathogenic Th17 cells in acetylcholine receptor antibody positive myasthenia gravis. Exp Neurol. 2020;325: 113146.PubMedCrossRef Yi JS, Russo MA, Raja S, Massey JM, Juel VC, Shin J, et al. Inhibition of the transcription factor ROR-γ reduces pathogenic Th17 cells in acetylcholine receptor antibody positive myasthenia gravis. Exp Neurol. 2020;325: 113146.PubMedCrossRef
62.
Zurück zum Zitat Ostlie NS, Karachunski PI, Wang W, Monfardini C, Kronenberg M, Conti-Fine BM. Transgenic expression of IL-10 in T cells facilitates development of experimental myasthenia gravis. J Immunol. 2001;166(8):4853–62.PubMedCrossRef Ostlie NS, Karachunski PI, Wang W, Monfardini C, Kronenberg M, Conti-Fine BM. Transgenic expression of IL-10 in T cells facilitates development of experimental myasthenia gravis. J Immunol. 2001;166(8):4853–62.PubMedCrossRef
63.
Zurück zum Zitat Hoffacker V, Schultz A, Tiesinga JJ, Gold R, Schalke B, Nix W, et al. Thymomas alter the T-cell subset composition in the blood: a potential mechanism for thymoma-associated autoimmune disease. Blood. 2000;96(12):3872–9.PubMedCrossRef Hoffacker V, Schultz A, Tiesinga JJ, Gold R, Schalke B, Nix W, et al. Thymomas alter the T-cell subset composition in the blood: a potential mechanism for thymoma-associated autoimmune disease. Blood. 2000;96(12):3872–9.PubMedCrossRef
64.
Zurück zum Zitat Tackenberg B, Nitschke M, Willcox N, Ziegler A, Nessler S, Schumm F, et al. CD45 isoform expression in autoimmune myasthenia gravis. Autoimmunity. 2003;36(2):117–21.PubMedCrossRef Tackenberg B, Nitschke M, Willcox N, Ziegler A, Nessler S, Schumm F, et al. CD45 isoform expression in autoimmune myasthenia gravis. Autoimmunity. 2003;36(2):117–21.PubMedCrossRef
65.
Zurück zum Zitat Zamecnik J, Vesely D, Jakubicka B, Simkova L, Pitha J, Schutzner J, et al. Muscle lymphocytic infiltrates in thymoma-associated myasthenia gravis are phenotypically different from those in polymyositis. Neuromuscular Disorders NMD. 2007;17(11–12):935–42.PubMedCrossRef Zamecnik J, Vesely D, Jakubicka B, Simkova L, Pitha J, Schutzner J, et al. Muscle lymphocytic infiltrates in thymoma-associated myasthenia gravis are phenotypically different from those in polymyositis. Neuromuscular Disorders NMD. 2007;17(11–12):935–42.PubMedCrossRef
66.
Zurück zum Zitat Tackenberg B, Schlegel K, Happel M, Eienbröker C, Gellert K, Oertel WH, et al. Expanded TCR Vbeta subsets of CD8(+) T-cells in late-onset myasthenia gravis: novel parallels with thymoma patients. J Neuroimmunol. 2009;216(1–2):85–91.PubMedCrossRef Tackenberg B, Schlegel K, Happel M, Eienbröker C, Gellert K, Oertel WH, et al. Expanded TCR Vbeta subsets of CD8(+) T-cells in late-onset myasthenia gravis: novel parallels with thymoma patients. J Neuroimmunol. 2009;216(1–2):85–91.PubMedCrossRef
67.
Zurück zum Zitat Teleshova N, Matusevicius D, Kivisäkk P, Mustafa M, Pirskanen R, Link H. Altered expression of costimulatory molecules in myasthenia gravis. Muscle Nerve. 2000;23(6):946–53.PubMedCrossRef Teleshova N, Matusevicius D, Kivisäkk P, Mustafa M, Pirskanen R, Link H. Altered expression of costimulatory molecules in myasthenia gravis. Muscle Nerve. 2000;23(6):946–53.PubMedCrossRef
68.
Zurück zum Zitat Krawczyk P, Adamczyk-Korbel M, Kieszko R, Korobowicz E, Milanowski J. Immunological system status and the appearance of respiratory system disturbances in thymectomized patients. Arch Immunol Ther Exp. 2007;55(1):49–56.CrossRef Krawczyk P, Adamczyk-Korbel M, Kieszko R, Korobowicz E, Milanowski J. Immunological system status and the appearance of respiratory system disturbances in thymectomized patients. Arch Immunol Ther Exp. 2007;55(1):49–56.CrossRef
69.
Zurück zum Zitat Suzuki Y, Onodera H, Tago H, Saito R, Ohuchi M, Shimizu M, et al. Altered expression of Th1-type chemokine receptor CXCR3 on CD4+ T cells in myasthenia gravis patients. J Neuroimmunol. 2006;172(1–2):166–74.PubMedCrossRef Suzuki Y, Onodera H, Tago H, Saito R, Ohuchi M, Shimizu M, et al. Altered expression of Th1-type chemokine receptor CXCR3 on CD4+ T cells in myasthenia gravis patients. J Neuroimmunol. 2006;172(1–2):166–74.PubMedCrossRef
70.
Zurück zum Zitat Matsumoto Y, Matsuo H, Sakuma H, Park IK, Tsukada Y, Kohyama K, et al. CDR3 spectratyping analysis of the TCR repertoire in myasthenia gravis. J Immunol. 2006;176(8):5100–7.PubMedCrossRef Matsumoto Y, Matsuo H, Sakuma H, Park IK, Tsukada Y, Kohyama K, et al. CDR3 spectratyping analysis of the TCR repertoire in myasthenia gravis. J Immunol. 2006;176(8):5100–7.PubMedCrossRef
71.
Zurück zum Zitat Nancy P, Berrih-Aknin S. Differential estrogen receptor expression in autoimmune myasthenia gravis. Endocrinology. 2005;146(5):2345–53.PubMedCrossRef Nancy P, Berrih-Aknin S. Differential estrogen receptor expression in autoimmune myasthenia gravis. Endocrinology. 2005;146(5):2345–53.PubMedCrossRef
72.
Zurück zum Zitat Feferman T, Maiti PK, Berrih-Aknin S, Bismuth J, Bidault J, Fuchs S, et al. Overexpression of IFN-induced protein 10 and its receptor CXCR3 in myasthenia gravis. J Immunol. 2005;174(9):5324–31.PubMedCrossRef Feferman T, Maiti PK, Berrih-Aknin S, Bismuth J, Bidault J, Fuchs S, et al. Overexpression of IFN-induced protein 10 and its receptor CXCR3 in myasthenia gravis. J Immunol. 2005;174(9):5324–31.PubMedCrossRef
73.
Zurück zum Zitat Mai W, Liu X, Fan Y, Liu H, Hong HY, Han R, et al. Up-regulated expression of Fas antigen in peripheral T cell subsets in patients with myasthenia gravis. Clin Invest Med. 2012;35(5):E294.PubMedCrossRef Mai W, Liu X, Fan Y, Liu H, Hong HY, Han R, et al. Up-regulated expression of Fas antigen in peripheral T cell subsets in patients with myasthenia gravis. Clin Invest Med. 2012;35(5):E294.PubMedCrossRef
75.
76.
Zurück zum Zitat Agle K, Vincent BG, Piper C, Belle L, Zhou V, Shlomchik W, et al. Bim regulates the survival and suppressive capability of CD8(+) FOXP3(+) regulatory T cells during murine GVHD. Blood. 2018;132(4):435–47.PubMedPubMedCentralCrossRef Agle K, Vincent BG, Piper C, Belle L, Zhou V, Shlomchik W, et al. Bim regulates the survival and suppressive capability of CD8(+) FOXP3(+) regulatory T cells during murine GVHD. Blood. 2018;132(4):435–47.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Beres AJ, Haribhai D, Chadwick AC, Gonyo PJ, Williams CB, Drobyski WR. CD8+ Foxp3+ regulatory T cells are induced during graft-versus-host disease and mitigate disease severity. J Immunol. 2012;189(1):464–74.PubMedCrossRef Beres AJ, Haribhai D, Chadwick AC, Gonyo PJ, Williams CB, Drobyski WR. CD8+ Foxp3+ regulatory T cells are induced during graft-versus-host disease and mitigate disease severity. J Immunol. 2012;189(1):464–74.PubMedCrossRef
78.
Zurück zum Zitat Robb RJ, Lineburg KE, Kuns RD, Wilson YA, Raffelt NC, Olver SD, et al. Identification and expansion of highly suppressive CD8(+)FoxP3(+) regulatory T cells after experimental allogeneic bone marrow transplantation. Blood. 2012;119(24):5898–908.PubMedCrossRef Robb RJ, Lineburg KE, Kuns RD, Wilson YA, Raffelt NC, Olver SD, et al. Identification and expansion of highly suppressive CD8(+)FoxP3(+) regulatory T cells after experimental allogeneic bone marrow transplantation. Blood. 2012;119(24):5898–908.PubMedCrossRef
79.
Zurück zum Zitat Sawamukai N, Satake A, Schmidt AM, Lamborn IT, Ojha P, Tanaka Y, et al. Cell-autonomous role of TGFβ and IL-2 receptors in CD4+ and CD8+ inducible regulatory T-cell generation during GVHD. Blood. 2012;119(23):5575–83.PubMedPubMedCentralCrossRef Sawamukai N, Satake A, Schmidt AM, Lamborn IT, Ojha P, Tanaka Y, et al. Cell-autonomous role of TGFβ and IL-2 receptors in CD4+ and CD8+ inducible regulatory T-cell generation during GVHD. Blood. 2012;119(23):5575–83.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Lerret NM, Houlihan JL, Kheradmand T, Pothoven KL, Zhang ZJ, Luo X. Donor-specific CD8+ Foxp3+ T cells protect skin allografts and facilitate induction of conventional CD4+ Foxp3+ regulatory T cells. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg. 2012;12(9):2335–47.CrossRef Lerret NM, Houlihan JL, Kheradmand T, Pothoven KL, Zhang ZJ, Luo X. Donor-specific CD8+ Foxp3+ T cells protect skin allografts and facilitate induction of conventional CD4+ Foxp3+ regulatory T cells. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg. 2012;12(9):2335–47.CrossRef
81.
Zurück zum Zitat Churlaud G, Pitoiset F, Jebbawi F, Lorenzon R, Bellier B, Rosenzwajg M, et al. Human and Mouse CD8(+)CD25(+)FOXP3(+) regulatory T cells at steady state and during interleukin-2 therapy. Front Immunol. 2015;6:171.PubMedPubMedCentralCrossRef Churlaud G, Pitoiset F, Jebbawi F, Lorenzon R, Bellier B, Rosenzwajg M, et al. Human and Mouse CD8(+)CD25(+)FOXP3(+) regulatory T cells at steady state and during interleukin-2 therapy. Front Immunol. 2015;6:171.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Zhong H, Liu Y, Xu Z, Liang P, Yang H, Zhang X, et al. TGF-β-induced CD8(+)CD103(+) regulatory T cells show potent therapeutic effect on chronic graft-versus-host disease lupus by suppressing B cells. Front Immunol. 2018;9:35.PubMedPubMedCentralCrossRef Zhong H, Liu Y, Xu Z, Liang P, Yang H, Zhang X, et al. TGF-β-induced CD8(+)CD103(+) regulatory T cells show potent therapeutic effect on chronic graft-versus-host disease lupus by suppressing B cells. Front Immunol. 2018;9:35.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Liu Y, Lan Q, Lu L, Chen M, Xia Z, Ma J, et al. Phenotypic and functional characteristic of a newly identified CD8+ Foxp3- CD103+ regulatory T cells. J Mol Cell Biol. 2014;6(1):81–92.PubMedCrossRef Liu Y, Lan Q, Lu L, Chen M, Xia Z, Ma J, et al. Phenotypic and functional characteristic of a newly identified CD8+ Foxp3- CD103+ regulatory T cells. J Mol Cell Biol. 2014;6(1):81–92.PubMedCrossRef
85.
Zurück zum Zitat Arosa FA, Esgalhado AJ, Padrão CA, Cardoso EM. Divide, conquer, and sense: CD8(+)CD28(-) T cells in perspective. Front Immunol. 2016;7:665.PubMed Arosa FA, Esgalhado AJ, Padrão CA, Cardoso EM. Divide, conquer, and sense: CD8(+)CD28(-) T cells in perspective. Front Immunol. 2016;7:665.PubMed
86.
Zurück zum Zitat Strioga M, Pasukoniene V, Characiejus D. CD8+ CD28- and CD8+ CD57+ T cells and their role in health and disease. Immunology. 2011;134(1):17–32.PubMedPubMedCentralCrossRef Strioga M, Pasukoniene V, Characiejus D. CD8+ CD28- and CD8+ CD57+ T cells and their role in health and disease. Immunology. 2011;134(1):17–32.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Dai H, Wan N, Zhang S, Moore Y, Wan F, Dai Z. Cutting edge: programmed death-1 defines CD8+CD122+ T cells as regulatory versus memory T cells. J Immunol. 2010;185(2):803–7.PubMedCrossRef Dai H, Wan N, Zhang S, Moore Y, Wan F, Dai Z. Cutting edge: programmed death-1 defines CD8+CD122+ T cells as regulatory versus memory T cells. J Immunol. 2010;185(2):803–7.PubMedCrossRef
89.
Zurück zum Zitat Okuno Y, Murakoshi A, Negita M, Akane K, Kojima S, Suzuki H. CD8+ CD122+ regulatory T cells contain clonally expanded cells with identical CDR3 sequences of the T-cell receptor β-chain. Immunology. 2013;139(3):309–17.PubMedPubMedCentralCrossRef Okuno Y, Murakoshi A, Negita M, Akane K, Kojima S, Suzuki H. CD8+ CD122+ regulatory T cells contain clonally expanded cells with identical CDR3 sequences of the T-cell receptor β-chain. Immunology. 2013;139(3):309–17.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Dai Z, Zhang S, Xie Q, Wu S, Su J, Li S, et al. Natural CD8+CD122+ T cells are more potent in suppression of allograft rejection than CD4+CD25+ regulatory T cells. Am J Transplant Off J Am Soc Transplant Am Soc Transpl Surg. 2014;14(1):39–48.CrossRef Dai Z, Zhang S, Xie Q, Wu S, Su J, Li S, et al. Natural CD8+CD122+ T cells are more potent in suppression of allograft rejection than CD4+CD25+ regulatory T cells. Am J Transplant Off J Am Soc Transplant Am Soc Transpl Surg. 2014;14(1):39–48.CrossRef
91.
Zurück zum Zitat Liu H, Qiu F, Wang Y, Zeng Q, Liu C, Chen Y, et al. CD8+CD122+PD-1+ tregs synergize with costimulatory blockade of CD40/CD154, but Not B7/CD28, to prolong murine allograft survival. Front Immunol. 2019;10:306.ADSPubMedPubMedCentralCrossRef Liu H, Qiu F, Wang Y, Zeng Q, Liu C, Chen Y, et al. CD8+CD122+PD-1+ tregs synergize with costimulatory blockade of CD40/CD154, but Not B7/CD28, to prolong murine allograft survival. Front Immunol. 2019;10:306.ADSPubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Hu D, Ikizawa K, Lu L, Sanchirico ME, Shinohara ML, Cantor H. Analysis of regulatory CD8 T cells in Qa-1-deficient mice. Nat Immunol. 2004;5:516.PubMedCrossRef Hu D, Ikizawa K, Lu L, Sanchirico ME, Shinohara ML, Cantor H. Analysis of regulatory CD8 T cells in Qa-1-deficient mice. Nat Immunol. 2004;5:516.PubMedCrossRef
94.
Zurück zum Zitat Lee YHIY, Rifa M, Shi Z, Isobe K, Suzuki H. Essential role of CD8+CD122+ regulatory T cells in the recovery from experimental autoimmune encephalomyelitis. J Immunol. 2008;180:825–32.PubMedCrossRef Lee YHIY, Rifa M, Shi Z, Isobe K, Suzuki H. Essential role of CD8+CD122+ regulatory T cells in the recovery from experimental autoimmune encephalomyelitis. J Immunol. 2008;180:825–32.PubMedCrossRef
95.
Zurück zum Zitat Yu PBR, Waldmann TA. IL-15-dependent CD8+ CD122+ T cells ameliorate experimental autoimmune encephalomyelitis by modulating IL-17 production by CD4+ T cells. Eur J Immunol. 2014;44:3330–41.PubMedPubMedCentralCrossRef Yu PBR, Waldmann TA. IL-15-dependent CD8+ CD122+ T cells ameliorate experimental autoimmune encephalomyelitis by modulating IL-17 production by CD4+ T cells. Eur J Immunol. 2014;44:3330–41.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Endharti ATOY, Shi Z, Misawa N, Toyokuni S, Ito M, et al. CD8+CD122+ regulatory T cells (Tregs) and CD4+ Tregs cooperatively prevent and cure CD4+ cell-induced colitis. J Immunol. 2011;186:41–52.PubMedCrossRef Endharti ATOY, Shi Z, Misawa N, Toyokuni S, Ito M, et al. CD8+CD122+ regulatory T cells (Tregs) and CD4+ Tregs cooperatively prevent and cure CD4+ cell-induced colitis. J Immunol. 2011;186:41–52.PubMedCrossRef
97.
Zurück zum Zitat Shimokawa C, Kato T, Takeuchi T, Ohshima N, Furuki T, Ohtsu Y, et al. CD8(+) regulatory T cells are critical in prevention of autoimmune-mediated diabetes. Nat Commun. 2020;11(1):1922.ADSPubMedPubMedCentralCrossRef Shimokawa C, Kato T, Takeuchi T, Ohshima N, Furuki T, Ohtsu Y, et al. CD8(+) regulatory T cells are critical in prevention of autoimmune-mediated diabetes. Nat Commun. 2020;11(1):1922.ADSPubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Varthaman A, Khallou-Laschet J, Clement M, Fornasa G, Kim HJ, Gaston AT, et al. Control of T cell reactivation by regulatory Qa-1-restricted CD8+ T cells. J Immunol. 2010;184(12):6585–91.PubMedCrossRef Varthaman A, Khallou-Laschet J, Clement M, Fornasa G, Kim HJ, Gaston AT, et al. Control of T cell reactivation by regulatory Qa-1-restricted CD8+ T cells. J Immunol. 2010;184(12):6585–91.PubMedCrossRef
99.
Zurück zum Zitat Leavenworth JW, Tang X, Kim HJ, Wang X, Cantor H. Amelioration of arthritis through mobilization of peptide-specific CD8+ regulatory T cells. J Clin Investig. 2013;123(3):1382–9.PubMedPubMedCentralCrossRef Leavenworth JW, Tang X, Kim HJ, Wang X, Cantor H. Amelioration of arthritis through mobilization of peptide-specific CD8+ regulatory T cells. J Clin Investig. 2013;123(3):1382–9.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Stocks BT, Wilson CS, Marshall AF, Hoopes EM, Moore DJ. Regulation of diabetogenic immunity by IL-15-activated regulatory CD8 T cells in type 1 diabetes. J Immunol. 2019;203(1):158–66.PubMedCrossRef Stocks BT, Wilson CS, Marshall AF, Hoopes EM, Moore DJ. Regulation of diabetogenic immunity by IL-15-activated regulatory CD8 T cells in type 1 diabetes. J Immunol. 2019;203(1):158–66.PubMedCrossRef
101.
Zurück zum Zitat Jiang H, Canfield SM, Gallagher MP, Jiang HH, Jiang Y, Zheng Z, et al. HLA-E-restricted regulatory CD8(+) T cells are involved in development and control of human autoimmune type 1 diabetes. J Clin Investig. 2010;120(10):3641–50.PubMedPubMedCentralCrossRef Jiang H, Canfield SM, Gallagher MP, Jiang HH, Jiang Y, Zheng Z, et al. HLA-E-restricted regulatory CD8(+) T cells are involved in development and control of human autoimmune type 1 diabetes. J Clin Investig. 2010;120(10):3641–50.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Holderried TA, Lang PA, Kim HJ, Cantor H. Genetic disruption of CD8+ Treg activity enhances the immune response to viral infection. Proc Natl Acad Sci USA. 2013;110(52):21089–94.ADSPubMedPubMedCentralCrossRef Holderried TA, Lang PA, Kim HJ, Cantor H. Genetic disruption of CD8+ Treg activity enhances the immune response to viral infection. Proc Natl Acad Sci USA. 2013;110(52):21089–94.ADSPubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Alvarez Arias DA, Kim HJ, Zhou P, Holderried TA, Wang X, Dranoff G, et al. Disruption of CD8+ Treg activity results in expansion of T follicular helper cells and enhanced antitumor immunity. Cancer Immunol Res. 2014;2(3):207–16.PubMedCrossRef Alvarez Arias DA, Kim HJ, Zhou P, Holderried TA, Wang X, Dranoff G, et al. Disruption of CD8+ Treg activity results in expansion of T follicular helper cells and enhanced antitumor immunity. Cancer Immunol Res. 2014;2(3):207–16.PubMedCrossRef
104.
Zurück zum Zitat Taghavie-Moghadam PL, Waseem TC, Hattler J, Glenn LM, Dobrian AD, Kaplan MH, et al. STAT4 regulates the CD8(+) regulatory T Cell/T follicular helper cell axis and promotes atherogenesis in insulin-resistant Ldlr(-/-) mice. J Immunol. 2017;199(10):3453–65.PubMedCrossRef Taghavie-Moghadam PL, Waseem TC, Hattler J, Glenn LM, Dobrian AD, Kaplan MH, et al. STAT4 regulates the CD8(+) regulatory T Cell/T follicular helper cell axis and promotes atherogenesis in insulin-resistant Ldlr(-/-) mice. J Immunol. 2017;199(10):3453–65.PubMedCrossRef
105.
Zurück zum Zitat Choi JY, Eskandari SK, Cai S, Sulkaj I, Assaker JP, Allos H, et al. Regulatory CD8 T cells that recognize Qa-1 expressed by CD4 T-helper cells inhibit rejection of heart allografts. Proc Natl Acad Sci USA. 2020;117(11):6042–6.ADSPubMedPubMedCentralCrossRef Choi JY, Eskandari SK, Cai S, Sulkaj I, Assaker JP, Allos H, et al. Regulatory CD8 T cells that recognize Qa-1 expressed by CD4 T-helper cells inhibit rejection of heart allografts. Proc Natl Acad Sci USA. 2020;117(11):6042–6.ADSPubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Li X, Xiao BG, Xi JY, Lu CZ, Lu JH. Decrease of CD4(+)CD25(high)Foxp3(+) regulatory T cells and elevation of CD19(+)BAFF-R(+) B cells and soluble ICAM-1 in myasthenia gravis. Clin Immunol. 2008;126(2):180–8.PubMedCrossRef Li X, Xiao BG, Xi JY, Lu CZ, Lu JH. Decrease of CD4(+)CD25(high)Foxp3(+) regulatory T cells and elevation of CD19(+)BAFF-R(+) B cells and soluble ICAM-1 in myasthenia gravis. Clin Immunol. 2008;126(2):180–8.PubMedCrossRef
107.
Zurück zum Zitat Ben-David H, Sharabi A, Dayan M, Sela M, Mozes E. The role of CD8+CD28 regulatory cells in suppressing myasthenia gravis-associated responses by a dual altered peptide ligand. Proc Natl Acad Sci USA. 2007;104(44):17459–64.ADSPubMedPubMedCentralCrossRef Ben-David H, Sharabi A, Dayan M, Sela M, Mozes E. The role of CD8+CD28 regulatory cells in suppressing myasthenia gravis-associated responses by a dual altered peptide ligand. Proc Natl Acad Sci USA. 2007;104(44):17459–64.ADSPubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Faurschou M, Jayne DR. Anti-B cell antibody therapies for inflammatory rheumatic diseases. Annu Rev Med. 2014;65:263–78.PubMedCrossRef Faurschou M, Jayne DR. Anti-B cell antibody therapies for inflammatory rheumatic diseases. Annu Rev Med. 2014;65:263–78.PubMedCrossRef
109.
Zurück zum Zitat Hauser SL, Waubant E, Arnold DL, Vollmer T, Antel J, Fox RJ, et al. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358(7):676–88.PubMedCrossRef Hauser SL, Waubant E, Arnold DL, Vollmer T, Antel J, Fox RJ, et al. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358(7):676–88.PubMedCrossRef
110.
Zurück zum Zitat Kappos L, Li D, Calabresi PA, O’Connor P, Bar-Or A, Barkhof F, et al. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. Lancet. 2011;378(9805):1779–87.PubMedCrossRef Kappos L, Li D, Calabresi PA, O’Connor P, Bar-Or A, Barkhof F, et al. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. Lancet. 2011;378(9805):1779–87.PubMedCrossRef
111.
Zurück zum Zitat Fernández O, Aladro Y, Arroyo R, Brieva L, Calles-Hernández MC, Carrascal P, et al. 12th Post-ECTRIMS meeting: review of the novelties from the 2019 ECTRIMS congress (II). Rev Neurol. 2020;70(11):417–29.PubMed Fernández O, Aladro Y, Arroyo R, Brieva L, Calles-Hernández MC, Carrascal P, et al. 12th Post-ECTRIMS meeting: review of the novelties from the 2019 ECTRIMS congress (II). Rev Neurol. 2020;70(11):417–29.PubMed
112.
Zurück zum Zitat Sorensen PS, Lisby S, Grove R, Derosier F, Shackelford S, Havrdova E, et al. Safety and efficacy of ofatumumab in relapsing-remitting multiple sclerosis: a phase 2 study. Neurology. 2014;82(7):573–81.PubMedCrossRef Sorensen PS, Lisby S, Grove R, Derosier F, Shackelford S, Havrdova E, et al. Safety and efficacy of ofatumumab in relapsing-remitting multiple sclerosis: a phase 2 study. Neurology. 2014;82(7):573–81.PubMedCrossRef
113.
Zurück zum Zitat Novikova DS, Popkova TV, Nasonov EL. The effect of anti-B-cell therapy on the development of atherosclerosis in patients with rheumatoid arthritis. Curr Pharm Des. 2012;18(11):1512–8.PubMedCrossRef Novikova DS, Popkova TV, Nasonov EL. The effect of anti-B-cell therapy on the development of atherosclerosis in patients with rheumatoid arthritis. Curr Pharm Des. 2012;18(11):1512–8.PubMedCrossRef
114.
Zurück zum Zitat Edwards JC, Szczepanski L, Szechinski J, Filipowicz-Sosnowska A, Emery P, Close DR, et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med. 2004;350(25):2572–81.PubMedCrossRef Edwards JC, Szczepanski L, Szechinski J, Filipowicz-Sosnowska A, Emery P, Close DR, et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med. 2004;350(25):2572–81.PubMedCrossRef
115.
Zurück zum Zitat Reddy V, Klein C, Isenberg DA, Glennie MJ, Cambridge G, Cragg MS, et al. Obinutuzumab induces superior B-cell cytotoxicity to rituximab in rheumatoid arthritis and systemic lupus erythematosus patient samples. Rheumatology. 2017;56(7):1227–37.PubMedPubMedCentralCrossRef Reddy V, Klein C, Isenberg DA, Glennie MJ, Cambridge G, Cragg MS, et al. Obinutuzumab induces superior B-cell cytotoxicity to rituximab in rheumatoid arthritis and systemic lupus erythematosus patient samples. Rheumatology. 2017;56(7):1227–37.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Kaegi C, Wuest B, Crowley C, Boyman O. systematic review of safety and efficacy of second- and third-generation CD20-targeting biologics in treating immune-mediated disorders. Front Immunol. 2021;12: 788830.PubMedCrossRef Kaegi C, Wuest B, Crowley C, Boyman O. systematic review of safety and efficacy of second- and third-generation CD20-targeting biologics in treating immune-mediated disorders. Front Immunol. 2021;12: 788830.PubMedCrossRef
118.
Zurück zum Zitat Reddy YN, Siedlecki AM, Francis JM. Breaking down the complement system: a review and update on novel therapies. Curr Opin Nephrol Hypertens. 2017;26(2):123–8.PubMed Reddy YN, Siedlecki AM, Francis JM. Breaking down the complement system: a review and update on novel therapies. Curr Opin Nephrol Hypertens. 2017;26(2):123–8.PubMed
119.
Zurück zum Zitat Klotz L, Wiendl H. Monoclonal antibodies in neuroinflammatory diseases. Expert Opin Biol Ther. 2013;13(6):831–46.PubMedCrossRef Klotz L, Wiendl H. Monoclonal antibodies in neuroinflammatory diseases. Expert Opin Biol Ther. 2013;13(6):831–46.PubMedCrossRef
120.
Zurück zum Zitat Meyer A, Lefevre G, Bierry G, Duval A, Ottaviani S, Meyer O, et al. In antisynthetase syndrome, ACPA are associated with severe and erosive arthritis: an overlapping rheumatoid arthritis and antisynthetase syndrome. Medicine. 2015;94(20): e523.PubMedPubMedCentralCrossRef Meyer A, Lefevre G, Bierry G, Duval A, Ottaviani S, Meyer O, et al. In antisynthetase syndrome, ACPA are associated with severe and erosive arthritis: an overlapping rheumatoid arthritis and antisynthetase syndrome. Medicine. 2015;94(20): e523.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Letaief H, Lukas C, Barnetche T, Gaujoux-Viala C, Combe B, Morel J. Efficacy and safety of biological DMARDs modulating B cells in primary Sjögren’s syndrome: Systematic review and meta-analysis. Joint Bone Spine. 2018;85(1):15–22.PubMedCrossRef Letaief H, Lukas C, Barnetche T, Gaujoux-Viala C, Combe B, Morel J. Efficacy and safety of biological DMARDs modulating B cells in primary Sjögren’s syndrome: Systematic review and meta-analysis. Joint Bone Spine. 2018;85(1):15–22.PubMedCrossRef
122.
Zurück zum Zitat Chen S, Liu Y, Shi G. Anti-CD20 antibody in primary Sjögren’s syndrome management. Curr Pharm Biotechnol. 2014;15(6):535–41.PubMedCrossRef Chen S, Liu Y, Shi G. Anti-CD20 antibody in primary Sjögren’s syndrome management. Curr Pharm Biotechnol. 2014;15(6):535–41.PubMedCrossRef
123.
Zurück zum Zitat Faurschou M, Hasselbalch HC, Nielsen OJ. Sustained remission of platelet counts following monoclonal anti-CD20 antibody therapy in two cases of idiopathic autoimmune thrombocytopenia and neutropenia. Eur J Haematol. 2001;66(6):408–11.PubMedCrossRef Faurschou M, Hasselbalch HC, Nielsen OJ. Sustained remission of platelet counts following monoclonal anti-CD20 antibody therapy in two cases of idiopathic autoimmune thrombocytopenia and neutropenia. Eur J Haematol. 2001;66(6):408–11.PubMedCrossRef
124.
Zurück zum Zitat Chen TX, Fan YT, Peng BW. Distinct mechanisms underlying therapeutic potentials of CD20 in neurological and neuromuscular disease. Pharmacol Ther. 2022;238: 108180.PubMedCrossRef Chen TX, Fan YT, Peng BW. Distinct mechanisms underlying therapeutic potentials of CD20 in neurological and neuromuscular disease. Pharmacol Ther. 2022;238: 108180.PubMedCrossRef
126.
Zurück zum Zitat Palanichamy A, Jahn S, Nickles D, Derstine M, Abounasr A, Hauser SL, et al. Rituximab efficiently depletes increased CD20-expressing T cells in multiple sclerosis patients. J Immunol. 2014;193(2):580–6.PubMedCrossRef Palanichamy A, Jahn S, Nickles D, Derstine M, Abounasr A, Hauser SL, et al. Rituximab efficiently depletes increased CD20-expressing T cells in multiple sclerosis patients. J Immunol. 2014;193(2):580–6.PubMedCrossRef
127.
Zurück zum Zitat Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol. 2006;59(6):880–92.PubMedCrossRef Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol. 2006;59(6):880–92.PubMedCrossRef
128.
Zurück zum Zitat von Büdingen HC, Bar-Or A, Zamvil SS. B cells in multiple sclerosis: connecting the dots. Curr Opin Immunol. 2011;23(6):713–20.CrossRef von Büdingen HC, Bar-Or A, Zamvil SS. B cells in multiple sclerosis: connecting the dots. Curr Opin Immunol. 2011;23(6):713–20.CrossRef
129.
Zurück zum Zitat Mohrmann RL, Arber DA. CD20-Positive peripheral T-cell lymphoma: report of a case after nodular sclerosis Hodgkin’s disease and review of the literature. Modern Pathol Off J United States Can Acad Pathol. 2000;13(11):1244–52.CrossRef Mohrmann RL, Arber DA. CD20-Positive peripheral T-cell lymphoma: report of a case after nodular sclerosis Hodgkin’s disease and review of the literature. Modern Pathol Off J United States Can Acad Pathol. 2000;13(11):1244–52.CrossRef
130.
Zurück zum Zitat Wilk E, Witte T, Marquardt N, Horvath T, Kalippke K, Scholz K, et al. Depletion of functionally active CD20+ T cells by rituximab treatment. Arthritis Rheum. 2009;60(12):3563–71.PubMedCrossRef Wilk E, Witte T, Marquardt N, Horvath T, Kalippke K, Scholz K, et al. Depletion of functionally active CD20+ T cells by rituximab treatment. Arthritis Rheum. 2009;60(12):3563–71.PubMedCrossRef
131.
Zurück zum Zitat Pan P, Pineda MA, Wang Y, Khan A, Nyirenda MH. Aberrant pro-inflammatory responses of CD20(+) T cells in experimental arthritis. Cell Immunol. 2023;387: 104717.PubMedCrossRef Pan P, Pineda MA, Wang Y, Khan A, Nyirenda MH. Aberrant pro-inflammatory responses of CD20(+) T cells in experimental arthritis. Cell Immunol. 2023;387: 104717.PubMedCrossRef
132.
Zurück zum Zitat Schuh E, Berer K, Mulazzani M, Feil K, Meinl I, Lahm H, et al. Features of human CD3+CD20+ T cells. J Immunol. 2016;197(4):1111–7.PubMedCrossRef Schuh E, Berer K, Mulazzani M, Feil K, Meinl I, Lahm H, et al. Features of human CD3+CD20+ T cells. J Immunol. 2016;197(4):1111–7.PubMedCrossRef
133.
Zurück zum Zitat Shinoda K, Li R, Rezk A, Mexhitaj I, Patterson KR, Kakara M, et al. Differential effects of anti-CD20 therapy on CD4 and CD8 T cells and implication of CD20-expressing CD8 T cells in MS disease activity. Proc Natl Acad Sci USA. 2023;120(3): e2207291120.PubMedPubMedCentralCrossRef Shinoda K, Li R, Rezk A, Mexhitaj I, Patterson KR, Kakara M, et al. Differential effects of anti-CD20 therapy on CD4 and CD8 T cells and implication of CD20-expressing CD8 T cells in MS disease activity. Proc Natl Acad Sci USA. 2023;120(3): e2207291120.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Gingele S, Jacobus TL, Konen FF, Hümmert MW, Sühs KW, Schwenkenbecher P, et al. Ocrelizumab depletes CD20+ T cells in multiple sclerosis patients. Cells. 2018;8(1):12.PubMedPubMedCentralCrossRef Gingele S, Jacobus TL, Konen FF, Hümmert MW, Sühs KW, Schwenkenbecher P, et al. Ocrelizumab depletes CD20+ T cells in multiple sclerosis patients. Cells. 2018;8(1):12.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat von Essen MR, Talbot J, Hansen RHH, Chow HH, Lundell H, Siebner HR, et al. Intrathecal CD8(+)CD20(+) T cells in primary progressive multiple sclerosis. Neurol Neuroimmunol Neuroinflam. 2023;10(5):200140.CrossRef von Essen MR, Talbot J, Hansen RHH, Chow HH, Lundell H, Siebner HR, et al. Intrathecal CD8(+)CD20(+) T cells in primary progressive multiple sclerosis. Neurol Neuroimmunol Neuroinflam. 2023;10(5):200140.CrossRef
136.
Zurück zum Zitat Ochs J, Nissimov N, Torke S, Freier M, Grondey K, Koch J, et al. Proinflammatory CD20(+) T cells contribute to CNS-directed autoimmunity. Sci Transl Med. 2022;14(638):eabi4632.PubMedCrossRef Ochs J, Nissimov N, Torke S, Freier M, Grondey K, Koch J, et al. Proinflammatory CD20(+) T cells contribute to CNS-directed autoimmunity. Sci Transl Med. 2022;14(638):eabi4632.PubMedCrossRef
137.
Zurück zum Zitat Marino M, Bartoccioni E, Alboini PE, Evoli A. Rituximab in myasthenia gravis: a “to be or not to be” inhibitor of T cell function. Ann N Y Acad Sci. 2018;1413(1):41–8.ADSPubMedCrossRef Marino M, Bartoccioni E, Alboini PE, Evoli A. Rituximab in myasthenia gravis: a “to be or not to be” inhibitor of T cell function. Ann N Y Acad Sci. 2018;1413(1):41–8.ADSPubMedCrossRef
139.
Zurück zum Zitat Boldrini VO, Quintiliano RPS, Silva LS, Damasceno A, Santos LMB, Farias AS. Cytotoxic profile of CD3+CD20+ T cells in progressive multiple sclerosis. Multiple Sclerosis Related Dis. 2021;52: 103013.CrossRef Boldrini VO, Quintiliano RPS, Silva LS, Damasceno A, Santos LMB, Farias AS. Cytotoxic profile of CD3+CD20+ T cells in progressive multiple sclerosis. Multiple Sclerosis Related Dis. 2021;52: 103013.CrossRef
140.
Zurück zum Zitat Shinoda K, Matsushita T, Nakamura Y, Masaki K, Sakai S, Nomiyama H, et al. Contribution of cortical lesions to cognitive impairment in Japanese patients with multiple sclerosis. Sci Rep. 2020;10(1):5228.ADSPubMedPubMedCentralCrossRef Shinoda K, Matsushita T, Nakamura Y, Masaki K, Sakai S, Nomiyama H, et al. Contribution of cortical lesions to cognitive impairment in Japanese patients with multiple sclerosis. Sci Rep. 2020;10(1):5228.ADSPubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Turner CN, Mullins GN, Hoyer KK. CXCR5(+)CD8 T cells: potential immunotherapy targets or drivers of immune-mediated adverse events? Front Med. 2022;9:1034764.CrossRef Turner CN, Mullins GN, Hoyer KK. CXCR5(+)CD8 T cells: potential immunotherapy targets or drivers of immune-mediated adverse events? Front Med. 2022;9:1034764.CrossRef
143.
Zurück zum Zitat Valentine KM, Mullins GN, Davalos OA, Seow LW, Hoyer KK. CD8 follicular T cells localize throughout the follicle during germinal center reactions and maintain cytolytic and helper properties. J Autoimmun. 2021;123: 102690.PubMedPubMedCentralCrossRef Valentine KM, Mullins GN, Davalos OA, Seow LW, Hoyer KK. CD8 follicular T cells localize throughout the follicle during germinal center reactions and maintain cytolytic and helper properties. J Autoimmun. 2021;123: 102690.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Haran KP, Hajduczki A, Pampusch MS, Mwakalundwa G, Vargas-Inchaustegui DA, Rakasz EG, et al. Simian immunodeficiency virus (SIV)-specific chimeric antigen receptor-T cells engineered to target B cell follicles and suppress SIV replication. Front Immunol. 2018;9:492.PubMedPubMedCentralCrossRef Haran KP, Hajduczki A, Pampusch MS, Mwakalundwa G, Vargas-Inchaustegui DA, Rakasz EG, et al. Simian immunodeficiency virus (SIV)-specific chimeric antigen receptor-T cells engineered to target B cell follicles and suppress SIV replication. Front Immunol. 2018;9:492.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Mylvaganam GH, Rios D, Abdelaal HM, Iyer S, Tharp G, Mavigner M, et al. Dynamics of SIV-specific CXCR5+ CD8 T cells during chronic SIV infection. Proc Natl Acad Sci USA. 2017;114(8):1976–81.ADSPubMedPubMedCentralCrossRef Mylvaganam GH, Rios D, Abdelaal HM, Iyer S, Tharp G, Mavigner M, et al. Dynamics of SIV-specific CXCR5+ CD8 T cells during chronic SIV infection. Proc Natl Acad Sci USA. 2017;114(8):1976–81.ADSPubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Cartwright EK, Pampusch MS, Rendahl AK, Berger EA, Coleman-Fuller N, Skinner PJ. HIV-specific CAR T cells with CD28 or 4–1BB signaling domains are phenotypically and functionally distinct and effective at suppressing HIV and simian immunodeficiency virus. ImmunoHorizons. 2022;6(10):693–704.PubMedCrossRef Cartwright EK, Pampusch MS, Rendahl AK, Berger EA, Coleman-Fuller N, Skinner PJ. HIV-specific CAR T cells with CD28 or 4–1BB signaling domains are phenotypically and functionally distinct and effective at suppressing HIV and simian immunodeficiency virus. ImmunoHorizons. 2022;6(10):693–704.PubMedCrossRef
147.
Zurück zum Zitat Huot N, Rascle P, Tchitchek N, Wimmer B, Passaes C, Contreras V, et al. Role of NKG2a/c+CD8+ T cells in pathogenic versus non-pathogenic SIV infections. iScience. 2021;24(4):102314.ADSPubMedPubMedCentralCrossRef Huot N, Rascle P, Tchitchek N, Wimmer B, Passaes C, Contreras V, et al. Role of NKG2a/c+CD8+ T cells in pathogenic versus non-pathogenic SIV infections. iScience. 2021;24(4):102314.ADSPubMedPubMedCentralCrossRef
149.
Zurück zum Zitat He R, Hou S, Liu C, Zhang A, Bai Q, Han M, et al. Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature. 2016;537(7620):412–28.ADSPubMedCrossRef He R, Hou S, Liu C, Zhang A, Bai Q, Han M, et al. Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature. 2016;537(7620):412–28.ADSPubMedCrossRef
150.
Zurück zum Zitat Mylvaganam GH, Velu V, Hong JJ, Sadagopal S, Kwa S, Basu R, et al. Diminished viral control during simian immunodeficiency virus infection is associated with aberrant PD-1hi CD4 T cell enrichment in the lymphoid follicles of the rectal mucosa. J Immunol. 2014;193(9):4527–36.PubMedCrossRef Mylvaganam GH, Velu V, Hong JJ, Sadagopal S, Kwa S, Basu R, et al. Diminished viral control during simian immunodeficiency virus infection is associated with aberrant PD-1hi CD4 T cell enrichment in the lymphoid follicles of the rectal mucosa. J Immunol. 2014;193(9):4527–36.PubMedCrossRef
151.
Zurück zum Zitat Perdomo-Celis F, Medina-Moreno S, Davis H, Bryant J, Taborda NA, Rugeles MT, et al. Characterization of CXCR5(+) CD8(+) T-cells in humanized NSG mice. Immunobiology. 2020;225(2): 151885.PubMedCrossRef Perdomo-Celis F, Medina-Moreno S, Davis H, Bryant J, Taborda NA, Rugeles MT, et al. Characterization of CXCR5(+) CD8(+) T-cells in humanized NSG mice. Immunobiology. 2020;225(2): 151885.PubMedCrossRef
152.
Zurück zum Zitat Perdomo-Celis F, Taborda NA, Rugeles MT. Circulating CXCR5-expressing CD8+ T-cells are major producers of IL-21 and associate with limited HIV replication. J Acquired Immune Deficien Syndrome. 2018;78(4):473–82.CrossRef Perdomo-Celis F, Taborda NA, Rugeles MT. Circulating CXCR5-expressing CD8+ T-cells are major producers of IL-21 and associate with limited HIV replication. J Acquired Immune Deficien Syndrome. 2018;78(4):473–82.CrossRef
153.
Zurück zum Zitat Perdomo-Celis F, Feria MG, Taborda NA, Rugeles MT. Induction of follicular-like CXCR5(+) CD8(+) T cells by TGF-β1/IL-23 Is limited during HIV infection. Viral Immunol. 2019;32(7):278–88.PubMedCrossRef Perdomo-Celis F, Feria MG, Taborda NA, Rugeles MT. Induction of follicular-like CXCR5(+) CD8(+) T cells by TGF-β1/IL-23 Is limited during HIV infection. Viral Immunol. 2019;32(7):278–88.PubMedCrossRef
154.
Zurück zum Zitat Starke CE, Vinton CL, Ladell K, McLaren JE, Ortiz AM, Mudd JC, et al. SIV-specific CD8+ T cells are clonotypically distinct across lymphoid and mucosal tissues. J Clin Investig. 2020;130(2):789–98.PubMedPubMedCentralCrossRef Starke CE, Vinton CL, Ladell K, McLaren JE, Ortiz AM, Mudd JC, et al. SIV-specific CD8+ T cells are clonotypically distinct across lymphoid and mucosal tissues. J Clin Investig. 2020;130(2):789–98.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Martínez LE, Ibarrondo J, Guo Y, Penichet ML, Epeldegui M. Follicular CD8+ T cells are elevated in HIV infection and induce PD-L1 on B cells. J Immunol. 2023;210(1):33–9.PubMedCrossRef Martínez LE, Ibarrondo J, Guo Y, Penichet ML, Epeldegui M. Follicular CD8+ T cells are elevated in HIV infection and induce PD-L1 on B cells. J Immunol. 2023;210(1):33–9.PubMedCrossRef
156.
Zurück zum Zitat Olivo A, Lécuroux C, Bitu M, Avettand-Fenoel V, Boufassa F, Essat A, et al. CXCR3 and CXCR5 are highly expressed in HIV-1-specific CD8 central memory T cells from infected patients. Eur J Immunol. 2021;51(8):2040–50.PubMedCrossRef Olivo A, Lécuroux C, Bitu M, Avettand-Fenoel V, Boufassa F, Essat A, et al. CXCR3 and CXCR5 are highly expressed in HIV-1-specific CD8 central memory T cells from infected patients. Eur J Immunol. 2021;51(8):2040–50.PubMedCrossRef
157.
Zurück zum Zitat Adams P, Iserentant G, Servais JY, Vandekerckhove L, Vanham G, Seguin-Devaux C. Cytotoxic CD8+ T cells expressing CXCR5 are detectable in HIV-1 elite controllers after prolonged in vitro peptide stimulation. Front Immunol. 2020;11: 622343.PubMedCrossRef Adams P, Iserentant G, Servais JY, Vandekerckhove L, Vanham G, Seguin-Devaux C. Cytotoxic CD8+ T cells expressing CXCR5 are detectable in HIV-1 elite controllers after prolonged in vitro peptide stimulation. Front Immunol. 2020;11: 622343.PubMedCrossRef
159.
Zurück zum Zitat Yang HG, Jiao YM, Huang HH, Zhang C, Zhang JY, Xu RN, et al. Transforming growth factor-β promotes the function of HIV-specific CXCR5(+) CD8 T cells. Microbiol Immunol. 2020;64(6):458–68.PubMedCrossRef Yang HG, Jiao YM, Huang HH, Zhang C, Zhang JY, Xu RN, et al. Transforming growth factor-β promotes the function of HIV-specific CXCR5(+) CD8 T cells. Microbiol Immunol. 2020;64(6):458–68.PubMedCrossRef
160.
Zurück zum Zitat Jiao YM, Yang HG, Huang HH, Tu B, Xing SJ, Mao L, et al. Dichotomous Roles of programmed cell death 1 on HIV-specific CXCR5(+) and CXCR5(-) CD8(+) T cells during chronic HIV infection. Front Immunol. 2017;8:1786.PubMedPubMedCentralCrossRef Jiao YM, Yang HG, Huang HH, Tu B, Xing SJ, Mao L, et al. Dichotomous Roles of programmed cell death 1 on HIV-specific CXCR5(+) and CXCR5(-) CD8(+) T cells during chronic HIV infection. Front Immunol. 2017;8:1786.PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat Roider J, Maehara T, Ngoepe A, Ramsuran D, Muenchhoff M, Adland E, et al. High-frequency, functional HIV-specific T-follicular helper and regulatory cells are present within germinal centers in children but not adults. Front Immunol. 2018;9:1975.PubMedPubMedCentralCrossRef Roider J, Maehara T, Ngoepe A, Ramsuran D, Muenchhoff M, Adland E, et al. High-frequency, functional HIV-specific T-follicular helper and regulatory cells are present within germinal centers in children but not adults. Front Immunol. 2018;9:1975.PubMedPubMedCentralCrossRef
162.
Zurück zum Zitat Munusamy Ponnan S, Thiruvengadam K, Kathirvel S, Shankar J, Rajaraman A, Mathaiyan M, et al. Elevated numbers of HIV-specific poly-functional CD8(+) T cells with stem cell-like and follicular homing phenotypes in HIV-exposed seronegative individuals. Front Immunol. 2021;12: 638144.PubMedPubMedCentralCrossRef Munusamy Ponnan S, Thiruvengadam K, Kathirvel S, Shankar J, Rajaraman A, Mathaiyan M, et al. Elevated numbers of HIV-specific poly-functional CD8(+) T cells with stem cell-like and follicular homing phenotypes in HIV-exposed seronegative individuals. Front Immunol. 2021;12: 638144.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat McCarty B, Mwamzuka M, Marshed F, Generoso M, Alvarez P, Ilmet T, et al. Low peripheral T follicular helper cells in perinatally HIV-infected children correlate with advancing HIV disease. Front Immunol. 2018;9:1901.PubMedPubMedCentralCrossRef McCarty B, Mwamzuka M, Marshed F, Generoso M, Alvarez P, Ilmet T, et al. Low peripheral T follicular helper cells in perinatally HIV-infected children correlate with advancing HIV disease. Front Immunol. 2018;9:1901.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Fardoos R, Nyquist SK, Asowata OE, Kazer SW, Singh A, Ngoepe A, et al. HIV specific CD8(+) T(RM)-like cells in tonsils express exhaustive signatures in the absence of natural HIV control. Front Immunol. 2022;13: 912038.PubMedPubMedCentralCrossRef Fardoos R, Nyquist SK, Asowata OE, Kazer SW, Singh A, Ngoepe A, et al. HIV specific CD8(+) T(RM)-like cells in tonsils express exhaustive signatures in the absence of natural HIV control. Front Immunol. 2022;13: 912038.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Collins DR, Hitschfel J, Urbach JM, Mylvaganam GH, Ly NL, Arshad U, et al. Cytolytic CD8(+) T cells infiltrate germinal centers to limit ongoing HIV replication in spontaneous controller lymph nodes. Sci Immunol. 2023;8(83):eade5872.PubMedPubMedCentralCrossRef Collins DR, Hitschfel J, Urbach JM, Mylvaganam GH, Ly NL, Arshad U, et al. Cytolytic CD8(+) T cells infiltrate germinal centers to limit ongoing HIV replication in spontaneous controller lymph nodes. Sci Immunol. 2023;8(83):eade5872.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Ponnan SM, Vidyavijayan KK, Thiruvengadam K, Hilda JN, Mathayan M, Murugavel KG, et al. Role of circulating T follicular helper cells and stem-like memory CD4(+) T cells in the pathogenesis of HIV-2 infection and disease progression. Front Immunol. 2021;12: 666388.PubMedPubMedCentralCrossRef Ponnan SM, Vidyavijayan KK, Thiruvengadam K, Hilda JN, Mathayan M, Murugavel KG, et al. Role of circulating T follicular helper cells and stem-like memory CD4(+) T cells in the pathogenesis of HIV-2 infection and disease progression. Front Immunol. 2021;12: 666388.PubMedPubMedCentralCrossRef
168.
Zurück zum Zitat Munusamy Ponnan S, Thiruvengadam K, Tellapragada C, Ambikan AT, Narayanan A, Kathirvel S, et al. Deciphering the role of mucosal immune responses and the cervicovaginal microbiome in resistance to HIV infection in HIV-exposed seronegative (HESN) women. Microbiol Spectr. 2021;9(2): e0047021.PubMedCrossRef Munusamy Ponnan S, Thiruvengadam K, Tellapragada C, Ambikan AT, Narayanan A, Kathirvel S, et al. Deciphering the role of mucosal immune responses and the cervicovaginal microbiome in resistance to HIV infection in HIV-exposed seronegative (HESN) women. Microbiol Spectr. 2021;9(2): e0047021.PubMedCrossRef
169.
Zurück zum Zitat George AF, Luo X, Neidleman J, Hoh R, Vohra P, Thomas R, et al. Deep phenotypic analysis of blood and lymphoid T and NK cells from HIV+ controllers and ART-suppressed individuals. Front Immunol. 2022;13: 803417.PubMedPubMedCentralCrossRef George AF, Luo X, Neidleman J, Hoh R, Vohra P, Thomas R, et al. Deep phenotypic analysis of blood and lymphoid T and NK cells from HIV+ controllers and ART-suppressed individuals. Front Immunol. 2022;13: 803417.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Yue FY, Lo C, Sakhdari A, Lee EY, Kovacs CM, Benko E, et al. HIV-specific IL-21 producing CD4+ T cells are induced in acute and chronic progressive HIV infection and are associated with relative viral control. J Immunol. 2010;185(1):498–506.PubMedCrossRef Yue FY, Lo C, Sakhdari A, Lee EY, Kovacs CM, Benko E, et al. HIV-specific IL-21 producing CD4+ T cells are induced in acute and chronic progressive HIV infection and are associated with relative viral control. J Immunol. 2010;185(1):498–506.PubMedCrossRef
171.
Zurück zum Zitat Im SJ, Konieczny BT, Hudson WH, Masopust D, Ahmed R. PD-1+ stemlike CD8 T cells are resident in lymphoid tissues during persistent LCMV infection. Proc Natl Acad Sci USA. 2020;117(8):4292–9.ADSPubMedPubMedCentralCrossRef Im SJ, Konieczny BT, Hudson WH, Masopust D, Ahmed R. PD-1+ stemlike CD8 T cells are resident in lymphoid tissues during persistent LCMV infection. Proc Natl Acad Sci USA. 2020;117(8):4292–9.ADSPubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Huang Z, Zak J, Pratumchai I, Shaabani N, Vartabedian VF, Nguyen N, et al. IL-27 promotes the expansion of self-renewing CD8(+) T cells in persistent viral infection. J Exp Med. 2019;216(8):1791–808.PubMedPubMedCentralCrossRef Huang Z, Zak J, Pratumchai I, Shaabani N, Vartabedian VF, Nguyen N, et al. IL-27 promotes the expansion of self-renewing CD8(+) T cells in persistent viral infection. J Exp Med. 2019;216(8):1791–808.PubMedPubMedCentralCrossRef
173.
Zurück zum Zitat Jiang H, Li L, Han J, Sun Z, Rong Y, Jin Y. CXCR5(+) CD8(+) T cells indirectly offer B cell help and are inversely correlated with viral load in chronic hepatitis B infection. DNA Cell Biol. 2017;36(4):321–7.PubMedCrossRef Jiang H, Li L, Han J, Sun Z, Rong Y, Jin Y. CXCR5(+) CD8(+) T cells indirectly offer B cell help and are inversely correlated with viral load in chronic hepatitis B infection. DNA Cell Biol. 2017;36(4):321–7.PubMedCrossRef
174.
Zurück zum Zitat Li Y, Tang L, Guo L, Chen C, Gu S, Zhou Y, et al. CXCL13-mediated recruitment of intrahepatic CXCR5(+)CD8(+) T cells favors viral control in chronic HBV infection. J Hepatol. 2020;72(3):420–30.PubMedCrossRef Li Y, Tang L, Guo L, Chen C, Gu S, Zhou Y, et al. CXCL13-mediated recruitment of intrahepatic CXCR5(+)CD8(+) T cells favors viral control in chronic HBV infection. J Hepatol. 2020;72(3):420–30.PubMedCrossRef
175.
Zurück zum Zitat Jin Y, Lang C, Tang J, Geng J, Song HK, Sun Z, et al. CXCR5(+)CD8(+) T cells could induce the death of tumor cells in HBV-related hepatocellular carcinoma. Int Immunopharmacol. 2017;53:42–8.PubMedCrossRef Jin Y, Lang C, Tang J, Geng J, Song HK, Sun Z, et al. CXCR5(+)CD8(+) T cells could induce the death of tumor cells in HBV-related hepatocellular carcinoma. Int Immunopharmacol. 2017;53:42–8.PubMedCrossRef
176.
Zurück zum Zitat Kumashie KG, Cebula M, Hagedorn C, Kreppel F, Pils MC, Koch-Nolte F, et al. Improved functionality of exhausted intrahepatic CXCR5+ CD8+ T cells contributes to chronic antigen clearance upon immunomodulation. Front Immunol. 2020;11: 592328.PubMedCrossRef Kumashie KG, Cebula M, Hagedorn C, Kreppel F, Pils MC, Koch-Nolte F, et al. Improved functionality of exhausted intrahepatic CXCR5+ CD8+ T cells contributes to chronic antigen clearance upon immunomodulation. Front Immunol. 2020;11: 592328.PubMedCrossRef
177.
Zurück zum Zitat Khanam A, Tang LSY, Kottilil S. Programmed death 1 expressing CD8(+) CXCR5(+) follicular T cells constitute effector rather than exhaustive phenotype in patients with chronic hepatitis B. Hepatology. 2022;75(3):690–708.PubMedCrossRef Khanam A, Tang LSY, Kottilil S. Programmed death 1 expressing CD8(+) CXCR5(+) follicular T cells constitute effector rather than exhaustive phenotype in patients with chronic hepatitis B. Hepatology. 2022;75(3):690–708.PubMedCrossRef
178.
Zurück zum Zitat Zhao H, Han Q, Yang A, Wang Y, Wang G, Lin A, et al. CpG-C ODN M362 as an immunoadjuvant for HBV therapeutic vaccine reverses the systemic tolerance against HBV. Int J Biol Sci. 2022;18(1):154–65.PubMedPubMedCentralCrossRef Zhao H, Han Q, Yang A, Wang Y, Wang G, Lin A, et al. CpG-C ODN M362 as an immunoadjuvant for HBV therapeutic vaccine reverses the systemic tolerance against HBV. Int J Biol Sci. 2022;18(1):154–65.PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Li X, Zhang Q, Zhang W, Ye G, Ma Y, Wen C, et al. Expanded circulating follicular dendritic cells facilitate immune responses in chronic HBV infection. J Transl Med. 2020;18(1):417.PubMedPubMedCentralCrossRef Li X, Zhang Q, Zhang W, Ye G, Ma Y, Wen C, et al. Expanded circulating follicular dendritic cells facilitate immune responses in chronic HBV infection. J Transl Med. 2020;18(1):417.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Tyllis TS, Fenix KA, Norton TS, Kara EE, McKenzie DR, David SC, et al. CXCR5(+)CD8(+) T cells shape antibody responses in vivo following protein immunisation and peripheral viral infection. Front Immunol. 2021;12: 626199.PubMedPubMedCentralCrossRef Tyllis TS, Fenix KA, Norton TS, Kara EE, McKenzie DR, David SC, et al. CXCR5(+)CD8(+) T cells shape antibody responses in vivo following protein immunisation and peripheral viral infection. Front Immunol. 2021;12: 626199.PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat Hoji A, Rinaldo CR Jr. Human CD8+ T cells specific for influenza A virus M1 display broad expression of maturation-associated phenotypic markers and chemokine receptors. Immunology. 2005;115(2):239–45.PubMedPubMedCentralCrossRef Hoji A, Rinaldo CR Jr. Human CD8+ T cells specific for influenza A virus M1 display broad expression of maturation-associated phenotypic markers and chemokine receptors. Immunology. 2005;115(2):239–45.PubMedPubMedCentralCrossRef
183.
Zurück zum Zitat Stanfield BA, Pahar B, Chouljenko VN, Veazey R, Kousoulas KG. Vaccination of rhesus macaques with the live-attenuated HSV-1 vaccine VC2 stimulates the proliferation of mucosal T cells and germinal center responses resulting in sustained production of highly neutralizing antibodies. Vaccine. 2017;35(4):536–43.PubMedCrossRef Stanfield BA, Pahar B, Chouljenko VN, Veazey R, Kousoulas KG. Vaccination of rhesus macaques with the live-attenuated HSV-1 vaccine VC2 stimulates the proliferation of mucosal T cells and germinal center responses resulting in sustained production of highly neutralizing antibodies. Vaccine. 2017;35(4):536–43.PubMedCrossRef
184.
Zurück zum Zitat Qiu L, Wang H, Yu Q, Liu J, Chen S, Zhao Z. Protective role of follicular CXCR5(+)CD8(+) T cells against dengue virus 2 infection. Int J Infect Dis Off Publ Int Soc Infect Dis. 2019;83:12–9.CrossRef Qiu L, Wang H, Yu Q, Liu J, Chen S, Zhao Z. Protective role of follicular CXCR5(+)CD8(+) T cells against dengue virus 2 infection. Int J Infect Dis Off Publ Int Soc Infect Dis. 2019;83:12–9.CrossRef
185.
Zurück zum Zitat Rivino L, Kumaran EA, Jovanovic V, Nadua K, Teo EW, Pang SW, et al. Differential targeting of viral components by CD4+ versus CD8+ T lymphocytes in dengue virus infection. J Virol. 2013;87(5):2693–706.PubMedPubMedCentralCrossRef Rivino L, Kumaran EA, Jovanovic V, Nadua K, Teo EW, Pang SW, et al. Differential targeting of viral components by CD4+ versus CD8+ T lymphocytes in dengue virus infection. J Virol. 2013;87(5):2693–706.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Zhou P, Gong F, Ji T, Cao C, Zheng T. Enriched CXCR3(+) CXCR5(+) CD8(+) T cells in SARS-CoV-2 infected and vaccinated individuals effectively respond to the antigen in recall. J Infect. 2023;86(5):497–9.PubMedPubMedCentralCrossRef Zhou P, Gong F, Ji T, Cao C, Zheng T. Enriched CXCR3(+) CXCR5(+) CD8(+) T cells in SARS-CoV-2 infected and vaccinated individuals effectively respond to the antigen in recall. J Infect. 2023;86(5):497–9.PubMedPubMedCentralCrossRef
189.
Zurück zum Zitat Esparcia-Pinedo L, Yarci-Carrión A, Mateo-Jiménez G, Ropero N, Gómez-Cabañas L, Lancho-Sánchez Á, et al. Development of an effective immune response in adults with down syndrome after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. Clin infect Dis Off Publ Infect Dis Soc Am. 2023;76(3):e155–62.CrossRef Esparcia-Pinedo L, Yarci-Carrión A, Mateo-Jiménez G, Ropero N, Gómez-Cabañas L, Lancho-Sánchez Á, et al. Development of an effective immune response in adults with down syndrome after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. Clin infect Dis Off Publ Infect Dis Soc Am. 2023;76(3):e155–62.CrossRef
190.
Zurück zum Zitat Yang J, Zhong M, Zhang E, Hong K, Yang Q, Zhou D, et al. Broad phenotypic alterations and potential dysfunction of lymphocytes in individuals clinically recovered from COVID-19. J Mol Cell Biol. 2021;13(3):197–209.PubMedPubMedCentralCrossRef Yang J, Zhong M, Zhang E, Hong K, Yang Q, Zhou D, et al. Broad phenotypic alterations and potential dysfunction of lymphocytes in individuals clinically recovered from COVID-19. J Mol Cell Biol. 2021;13(3):197–209.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Chatterjee B, Deng Y, Holler A, Nunez N, Azzi T, Vanoaica LD, et al. CD8+ T cells retain protective functions despite sustained inhibitory receptor expression during Epstein-Barr virus infection in vivo. PLoS Pathog. 2019;15(5): e1007748.PubMedPubMedCentralCrossRef Chatterjee B, Deng Y, Holler A, Nunez N, Azzi T, Vanoaica LD, et al. CD8+ T cells retain protective functions despite sustained inhibitory receptor expression during Epstein-Barr virus infection in vivo. PLoS Pathog. 2019;15(5): e1007748.PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat Jifu E, Yan F, Kang Z, Zhu L, Xing J, Enda Y. CD8(+)CXCR5(+) T cells in tumor-draining lymph nodes are highly activated and predict better prognosis in colorectal cancer. Human Immunol. 2018;79(6):446–52.CrossRef Jifu E, Yan F, Kang Z, Zhu L, Xing J, Enda Y. CD8(+)CXCR5(+) T cells in tumor-draining lymph nodes are highly activated and predict better prognosis in colorectal cancer. Human Immunol. 2018;79(6):446–52.CrossRef
193.
Zurück zum Zitat Shen J, Luo X, Wu Q, Huang J, Xiao G, Wang L, et al. A subset of CXCR5(+)CD8(+) T cells in the germinal centers from human tonsils and lymph nodes help B cells produce immunoglobulins. Front Immunol. 2018;9:2287.PubMedPubMedCentralCrossRef Shen J, Luo X, Wu Q, Huang J, Xiao G, Wang L, et al. A subset of CXCR5(+)CD8(+) T cells in the germinal centers from human tonsils and lymph nodes help B cells produce immunoglobulins. Front Immunol. 2018;9:2287.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Xing J, Zhang C, Yang X, Wang S, Wang Z, Li X, et al. CXCR5(+)CD8(+) T cells infiltrate the colorectal tumors and nearby lymph nodes, and are associated with enhanced IgG response in B cells. Exp Cell Res. 2017;356(1):57–63.PubMed Xing J, Zhang C, Yang X, Wang S, Wang Z, Li X, et al. CXCR5(+)CD8(+) T cells infiltrate the colorectal tumors and nearby lymph nodes, and are associated with enhanced IgG response in B cells. Exp Cell Res. 2017;356(1):57–63.PubMed
195.
Zurück zum Zitat Brummelman J, Mazza EMC, Alvisi G, Colombo FS, Grilli A, Mikulak J, et al. High-dimensional single cell analysis identifies stem-like cytotoxic CD8(+) T cells infiltrating human tumors. J Exp Med. 2018;215(10):2520–35.PubMedPubMedCentralCrossRef Brummelman J, Mazza EMC, Alvisi G, Colombo FS, Grilli A, Mikulak J, et al. High-dimensional single cell analysis identifies stem-like cytotoxic CD8(+) T cells infiltrating human tumors. J Exp Med. 2018;215(10):2520–35.PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat Ye L, Li Y, Tang H, Liu W, Chen Y, Dai T, et al. CD8+CXCR5+T cells infiltrating hepatocellular carcinomas are activated and predictive of a better prognosis. Aging. 2019;11(20):8879–91.PubMedPubMedCentralCrossRef Ye L, Li Y, Tang H, Liu W, Chen Y, Dai T, et al. CD8+CXCR5+T cells infiltrating hepatocellular carcinomas are activated and predictive of a better prognosis. Aging. 2019;11(20):8879–91.PubMedPubMedCentralCrossRef
197.
Zurück zum Zitat Hofland T, Martens AWJ, van Bruggen JAC, de Boer R, Schetters S, Remmerswaal EBM, et al. Human CXCR5(+) PD-1(+) CD8 T cells in healthy individuals and patients with hematologic malignancies. Eur J Immunol. 2021;51(3):703–13.PubMedCrossRef Hofland T, Martens AWJ, van Bruggen JAC, de Boer R, Schetters S, Remmerswaal EBM, et al. Human CXCR5(+) PD-1(+) CD8 T cells in healthy individuals and patients with hematologic malignancies. Eur J Immunol. 2021;51(3):703–13.PubMedCrossRef
198.
Zurück zum Zitat Bai M, Zheng Y, Liu H, Su B, Zhan Y, He H. CXCR5(+) CD8(+) T cells potently infiltrate pancreatic tumors and present high functionality. Exp Cell Res. 2017;361(1):39–45.PubMedCrossRef Bai M, Zheng Y, Liu H, Su B, Zhan Y, He H. CXCR5(+) CD8(+) T cells potently infiltrate pancreatic tumors and present high functionality. Exp Cell Res. 2017;361(1):39–45.PubMedCrossRef
199.
Zurück zum Zitat Wang J, Li R, Cao Y, Gu Y, Fang H, Fei Y, et al. Intratumoral CXCR5(+)CD8(+)T associates with favorable clinical outcomes and immunogenic contexture in gastric cancer. Nat Commun. 2021;12(1):3080.ADSPubMedPubMedCentralCrossRef Wang J, Li R, Cao Y, Gu Y, Fang H, Fei Y, et al. Intratumoral CXCR5(+)CD8(+)T associates with favorable clinical outcomes and immunogenic contexture in gastric cancer. Nat Commun. 2021;12(1):3080.ADSPubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Bassez A, Vos H, Van Dyck L, Floris G, Arijs I, Desmedt C, et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat Med. 2021;27(5):820–32.PubMedCrossRef Bassez A, Vos H, Van Dyck L, Floris G, Arijs I, Desmedt C, et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat Med. 2021;27(5):820–32.PubMedCrossRef
201.
Zurück zum Zitat Zhou Y, Guo L, Sun H, Xu J, Ba T. CXCR5(+) CD8 T cells displayed higher activation potential despite high PD-1 expression, in tumor-involved lymph nodes from patients with thyroid cancer. Int Immunopharmacol. 2018;62:114–9.PubMedCrossRef Zhou Y, Guo L, Sun H, Xu J, Ba T. CXCR5(+) CD8 T cells displayed higher activation potential despite high PD-1 expression, in tumor-involved lymph nodes from patients with thyroid cancer. Int Immunopharmacol. 2018;62:114–9.PubMedCrossRef
203.
Zurück zum Zitat Tang J, Zha J, Guo X, Shi P, Xu B. CXCR5(+)CD8(+) T cells present elevated capacity in mediating cytotoxicity toward autologous tumor cells through interleukin 10 in diffuse large B-cell lymphoma. Int Immunopharmacol. 2017;50:146–51.PubMedCrossRef Tang J, Zha J, Guo X, Shi P, Xu B. CXCR5(+)CD8(+) T cells present elevated capacity in mediating cytotoxicity toward autologous tumor cells through interleukin 10 in diffuse large B-cell lymphoma. Int Immunopharmacol. 2017;50:146–51.PubMedCrossRef
204.
Zurück zum Zitat Shen Y, Qu QX, Jin MN, Chen C. Investigating the role of circulating CXCR5-expressing CD8+ T-cells as a biomarker for bacterial infection in subjects with pneumonia. Respir Res. 2019;20(1):54.PubMedPubMedCentralCrossRef Shen Y, Qu QX, Jin MN, Chen C. Investigating the role of circulating CXCR5-expressing CD8+ T-cells as a biomarker for bacterial infection in subjects with pneumonia. Respir Res. 2019;20(1):54.PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Diupotex M, Zamora-Chimal J, Gajón JA, Bonifaz LC, Becker I. CXCR5 and TIM-3 expressions define distinct exhausted T cell subsets in experimental cutaneous infection with Leishmania mexicana. Front Immunol. 2023;14:1231836.PubMedPubMedCentralCrossRef Diupotex M, Zamora-Chimal J, Gajón JA, Bonifaz LC, Becker I. CXCR5 and TIM-3 expressions define distinct exhausted T cell subsets in experimental cutaneous infection with Leishmania mexicana. Front Immunol. 2023;14:1231836.PubMedPubMedCentralCrossRef
206.
Zurück zum Zitat de Lollo C, de Moraes VD, da Silva Oliveira LM, de Oliveira TT, Carneiro-Sampaio M, Jacob CM, et al. Impaired CD8(+) T cell responses upon Toll-like receptor activation in common variable immunodeficiency. J Transl Med. 2016;14(1):138.PubMedPubMedCentralCrossRef de Lollo C, de Moraes VD, da Silva Oliveira LM, de Oliveira TT, Carneiro-Sampaio M, Jacob CM, et al. Impaired CD8(+) T cell responses upon Toll-like receptor activation in common variable immunodeficiency. J Transl Med. 2016;14(1):138.PubMedPubMedCentralCrossRef
207.
Zurück zum Zitat Higashioka K, Yoshimura M, Sakuragi T, Ayano M, Kimoto Y, Mitoma H, et al. Human PD-1(hi)CD8(+) T cells are a cellular source of IL-21 in rheumatoid arthritis. Front Immunol. 2021;12: 654623.PubMedPubMedCentralCrossRef Higashioka K, Yoshimura M, Sakuragi T, Ayano M, Kimoto Y, Mitoma H, et al. Human PD-1(hi)CD8(+) T cells are a cellular source of IL-21 in rheumatoid arthritis. Front Immunol. 2021;12: 654623.PubMedPubMedCentralCrossRef
208.
Zurück zum Zitat Zhai X, Wang Y, Guo H, Liang Z, Feng M, Wu Y, et al. Altered levels of circulating CD8(+)CXCR5(+)PD-1(+)T follicular cytotoxic cells in primary Sjögren’s syndrome. Clin Rheumatol. 2022;41(6):1697–708.PubMedCrossRef Zhai X, Wang Y, Guo H, Liang Z, Feng M, Wu Y, et al. Altered levels of circulating CD8(+)CXCR5(+)PD-1(+)T follicular cytotoxic cells in primary Sjögren’s syndrome. Clin Rheumatol. 2022;41(6):1697–708.PubMedCrossRef
209.
Zurück zum Zitat Valentine KM, Davini D, Lawrence TJ, Mullins GN, Manansala M, Al-Kuhlani M, et al. CD8 follicular T cells promote B cell antibody class switch in autoimmune disease. J Immunol. 2018;201(1):31–40.PubMedCrossRef Valentine KM, Davini D, Lawrence TJ, Mullins GN, Manansala M, Al-Kuhlani M, et al. CD8 follicular T cells promote B cell antibody class switch in autoimmune disease. J Immunol. 2018;201(1):31–40.PubMedCrossRef
210.
Zurück zum Zitat Huber JE, Chang Y, Meinl I, Kümpfel T, Meinl E, Baumjohann D. Fingolimod profoundly reduces frequencies and alters subset composition of circulating T follicular helper cells in multiple sclerosis patients. J Immunol. 2020;204(5):1101–10.PubMedCrossRef Huber JE, Chang Y, Meinl I, Kümpfel T, Meinl E, Baumjohann D. Fingolimod profoundly reduces frequencies and alters subset composition of circulating T follicular helper cells in multiple sclerosis patients. J Immunol. 2020;204(5):1101–10.PubMedCrossRef
211.
Zurück zum Zitat Anang DC, Ramwadhdoebe TH, Hähnlein JS, van Kuijk B, Smits N, van Lienden KP, et al. Increased frequency of CD4(+) follicular helper T and CD8(+) follicular T cells in human lymph node biopsies during the earliest stages of rheumatoid arthritis. Cells. 2022;11(7):104.CrossRef Anang DC, Ramwadhdoebe TH, Hähnlein JS, van Kuijk B, Smits N, van Lienden KP, et al. Increased frequency of CD4(+) follicular helper T and CD8(+) follicular T cells in human lymph node biopsies during the earliest stages of rheumatoid arthritis. Cells. 2022;11(7):104.CrossRef
212.
Zurück zum Zitat Hinrichs AC, Kruize AA, Leavis HL, van Roon JAG. In patients with primary Sjögren’s syndrome innate-like MAIT cells display upregulated IL-7R, IFN-γ, and IL-21 expression and have increased proportions of CCR9 and CXCR5-expressing cells. Front Immunol. 2022;13:1017157.PubMedPubMedCentralCrossRef Hinrichs AC, Kruize AA, Leavis HL, van Roon JAG. In patients with primary Sjögren’s syndrome innate-like MAIT cells display upregulated IL-7R, IFN-γ, and IL-21 expression and have increased proportions of CCR9 and CXCR5-expressing cells. Front Immunol. 2022;13:1017157.PubMedPubMedCentralCrossRef
213.
Zurück zum Zitat Longbrake EE, Cantoni C, Chahin S, Cignarella F, Cross AH, Piccio L. Dimethyl fumarate induces changes in B- and T-lymphocyte function independent of the effects on absolute lymphocyte count. Multiple Sclerosis. 2018;24(6):728–38.PubMedCrossRef Longbrake EE, Cantoni C, Chahin S, Cignarella F, Cross AH, Piccio L. Dimethyl fumarate induces changes in B- and T-lymphocyte function independent of the effects on absolute lymphocyte count. Multiple Sclerosis. 2018;24(6):728–38.PubMedCrossRef
214.
Zurück zum Zitat Beura LK, Scott MC, Pierson MJ, Joag V, Wijeyesinghe S, Semler MR, et al. Novel lymphocytic choriomeningitis virus strain sustains abundant exhausted progenitor CD8 T cells without systemic viremia. J immunol. 2022;209(9):1691–702.PubMedCrossRef Beura LK, Scott MC, Pierson MJ, Joag V, Wijeyesinghe S, Semler MR, et al. Novel lymphocytic choriomeningitis virus strain sustains abundant exhausted progenitor CD8 T cells without systemic viremia. J immunol. 2022;209(9):1691–702.PubMedCrossRef
Metadaten
Titel
An angel or a devil? Current view on the role of CD8+ T cells in the pathogenesis of myasthenia gravis
verfasst von
Yong Peng
Huan Yang
Quan Chen
Hong Jin
Ya-hui Xue
Miao-qiao Du
Shu Liu
Shun-yu Yao
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2024
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-024-04965-7

Weitere Artikel der Ausgabe 1/2024

Journal of Translational Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.