- Research
- Open access
- Published:
Semaphorin 5A promotes Th17 differentiation via PI3K-Akt-mTOR in systemic lupus erythematosus
Arthritis Research & Therapy volume 26, Article number: 204 (2024)
Abstract
Background
Previously, we reported that serum Semaphorin 5 A (Sema5A) levels were increased in systemic lupus erythematosus (SLE) patients compared with healthy controls (HC), and elevated Sema5A correlated with disease activity and lupus nephritis in SLE patients. In this study, we aimed to further understand the role of Sema5A in promoting Th17 cells differentiation in SLE.
Methods
Sema5A, interferon gamma (IFN-γ), interleukin 4 (IL-4), interleukin 17 A (IL-17 A) and interleukin 10 (IL-10) were measured by Enzyme Linked Immunosorbent Assay (ELISA). RNA and protein were isolated from peripheral blood mononuclear cells (PBMCs) in SLE patients and HC. Expression of PlexinA1 and PlexinB3 were measured by quantitative RT-PCR (qRT-PCR) and Western Blot. Th cell subsets were detected by flow cytometry. Treatment with recombinant human Sema5A (rhSema5A) and small interfering RNA (siRNA) were employed to examine the in vitro effect of Sema5A in CD4+T cell differentiation in SLE patients.
Results
IL-17 A elevated in SLE patients and positively correlated with Sema5A. PlexinA1 was upregulated and mainly expressed in CD4+ T cells of SLE; Sema5A treatment induced the differentiation of Th17 cells, while did not affect the Th1 and Th2 skewing. These effects were associated with an upregulation of the transcription factor RORγt by Th17 cells, but not T-bet or GATA3 in Th1 and Th2 cells, respectively. Knock down PlexinA1 regulates IL-17 A production by CD4+T cells. Functional assays showed that Sema5A-PlexinA1 axis promoted Th17 cells differentiation via PI3K/Akt/mTOR signaling.
Conclusions
These findings demonstrated that Sema5A-PlexinA1 axis acts as a key mediator on Th17 differentiation, suggesting that Sema5A might be a novel therapeutic target in SLE.
Introduction
Systemic Lupus Erythematosus (SLE) is a systemic autoimmune disease characterized by the production of excessive anti-nuclear antibodies (ANAs), immune complex formation, and complement activation leading to target organ damage [1]. The clinical features of SLE are diverse, varying from mild fatigue, rash, and arthralgia to severe organ dysfunction and even life-threatening damage [2]. Multiple abnormalities of the immune system contribute to the pathogenesis of SLE [3, 4]. Among these abnormalities, the skewing of CD4+T cell differentiation plays a crucial role. In different microenvironments, CD4+T cells can differentiate into different helper T cells (Th) or regulatory T cells (Treg) after antigen activation [5, 6]. The Th cells can mediate B cell differentiation and autoantibodies secretion, and produce inflammatory cytokines, mediating different immune cells activation and inflammatory damage. Th17 cells, which mainly secrete IL-17, can promote the recruitment of neutrophils in inflammatory tissues. T cells secreting IL-17 have been reported infiltrated in the kidney tissues of lupus mice and lupus nephritis patients and are related to the degree of kidney damage, suggesting that Th17 / IL-17 plays a critical role in organ damage in SLE patients [7].
Semaphorins (Sema), are a family of molecules initially found to be involved in the guidance and repair of neuronal axon growth [8]. Its molecular structure contains a “Sema” domain, which can be divided into I-VIII categories. Various studies have found that Sema molecules are involved in physiological and pathological processes such as immune cell migration, immune function, cardiovascular regulation, renal homeostasis, and tumor metastasis [9]. “Immune Sema molecules” are Sema molecules with immunomodulatory function, which play important role in the normal immune response as well as pathological autoimmune processes [10]. Semaphorin 5 A (Sema5A) was originally found to be expressed in oligodendrocytes in the nervous system and inhibits axonal growth in retinal ganglions [11]. Studies have reported that its receptors are PlexinA1 and PlexinB3 [12, 13]. The altered expression and functions of Sema5A in autoimmune diseases like immune thrombocytopenic purpura, Hashimoto’s thyroiditis, rheumatoid arthritis, and chronic spontaneous urticaria have been reported recently. The level of soluble semaphorin 5 A (sSema5A) in serum is positively correlated with serum IFN-γ to IL-4 ratio in ITP while positively correlated with IL-17 A mRNA level in PBMCs of Hashimoto’s thyroiditis patients [14, 15]. In the study of chronic urticaria, Sema5A was found to be co-localized with skin basophils and was associated with the expression of IL-17 [16].
Previously, we reported that serum Sema5A levels were increased significantly in SLE patients compared with healthy controls. A disintegrin and metalloproteinases 17(ADAM17) might be involved in the release of secreted Sema5A. Besides, elevated serum Sema5A in SLE patients correlated with disease activity and is involved in kidney and blood system damage [17]. According to its correlation with Th cell cytokines, it is reasonable to predict that Sema5A may also be involved in the pathophysiology of SLE [14,15,16]. In this study, our findings indicate that Sema5A plays a direct role in promoting Th17 cell differentiation via PI3K/Akt/mTOR signaling, suggesting that Sema5A might be a novel therapeutic target in SLE.
Materials and methods
Patients and controls
Blood from 33 SLE patients and 30 sex- and age-matched healthy controls was obtained from the Second Affiliated Hospital of Zhejiang University School of Medicine. All subjects provided written informed consent before enrollment. Patients fulfilled the classification criteria of SLE revised by the American College of Rheumatology in 1997 [18]. Healthy controls were recruited from healthy staff members of the hospital. This study was approved by the Ethics Committee of the Second Affiliated Hospital of Zhejiang University School of Medicine (the approval protocol No: 2020433).
Peripheral blood mononuclear cells (PBMCs) separation and stimulation
PBMCs were separated from SLE patients and healthy controls using the density gradient centrifugation method. In some experiments, separated PBMCs were resuspended in MACS® Separation Buffer (130-091-221, Miltenyi Biotec) for subsets separation. In some experiments, separated PBMCs were resuspended in Roswell Park Memorial Institute (RPMI) 1640 medium (Gibco) before stimulation. The 48-well cell culture plate was coated with anti-human CD3 antibody (1 µg/mL, #300302, Biolegend) and cultured overnight at 4 °C, the coating solution was discarded before use. 1 × 106 PBMCs were seeded in the pre-coated culture plate, and soluble anti-human CD28 antibody (1 µg/mL, #302902, Biolegend) was added at the same time. Recombinant human Sema5A (#C499, Novoprotein) was added at a concentration of 1 µg/mL in the experimental group, and the same volume of sterile PBS was added in the control group. The cells were then cultured in an incubator at 5% CO2 and 37 °C for 48 h.For intracellular cytokine staining, phorbol 12-myristate 13-acetate (PMA, 1 µg/ml), ionomycin (50 ng/ml) and BD GolgiStop (BD Biosciences) were added for the final 4 h of stimulation. For transcriptional and cytokine analysis, cells and cell-free tissue culture supernatants were harvested for the following experiments.
Isolation of subsets from PBMCs
CD4+T cell, CD8+T cell, CD19+B cell, and CD14+ monocytes were separated in sequence from unstimulated PBMCs using immunomagnetic MicroBeads (#130-045-101, 130-045-201, 130-050-301, 130-050-201; Miltenyi Biotec). Isolated cells were saved in RPMI 1640 medium for the following experiments.
CD4+T cell differentiation
Purified CD4+ T cells (1 × 106 cells/ml) were polarized to Th0 cells using anti-human CD3 antibody pre-coated 96-well plates and soluble anti-human CD28 antibody. Th17 cell differentiation conditions were: IL-1β (10ng/ml, MCE); IL-6 (10ng/ml, MCE); TGF-β (5ng/ml, MCE); IL-23 (10ng/ml, MCE); anti-IL-4 (3 µg/ml, Biolegend); and anti-IFN-γ (3 µg/ml, Biolegend). Cells were cultured in the presence or absence of Sema5A (1 µg/mL) for 5 days. Cell-free tissue culture supernatants were harvested for cytokine analysis.
Small interfering RNA transfection
CD4+ T cells were activated with human T-Activator CD3/CD28 for 48 hours and polarized to Th0 cells before transfection. In order to confirm Sema5A promotes Th17 differentiation via connecting with plexin A1, Th0 cells were transfected with PlexinA1-specific or scramble (NC) non-targeting small interfering RNAs (siRNAs) using the GP Transfected-Mate reagent (Gene Pharma) for 48 hours, then cultured in Th17 differentiation condition with or without Sema5A (1µg/ml) for 5 days. Cell-free supernatants were collected for IL-17 analysis. In the signaling research, after transfection, Th0 cells were incubated with Sema5A(1µg/ml) for 24 hours and added Th17 cell differentiation conditions for the final 1 hour of stimulation. The sequence of the siRNA-PLXNA1 was as follows: 5’- GCAGUACUGACAACGUCAATT-3’.
Treatment with small-molecule inhibitor
Activated CD4+ T cells were treated with small-molecule inhibitors of PI3K-AKT-mTOR signaling to detect the effects of Sema5A after inhibition of this pathway. They are PI3K inhibitor, pictilisib (GDC-0941, HY-50094; MCE) 10 µM, AKT inhibitor, MK-2206 (HY-108232; MCE) 5 µM, and mTOR inhibitor, temsirolimus (CCI-779, HY-50910; MCE) 100 nM.
Enzyme-linked immunosorbent assay (ELISA)
Sema5A (Jiyinmei, Wuhan, P.R. China), IFN-γ, IL-4, IL-17 and IL-10 (Biolegend) were measured by enzyme-linked immunosorbent assay in cell-free supernatants and plasma from SLE patients and healthy controls, according to the manufacturer’s instructions.
Real-time quantitative PCR
Complementary DNA was synthesized from isolated RNA according to the instructions indicated in PrimeScript™ RT reagent Kit with gDNA Eraser (# RR047A, Takara, Japan). SYBR Green qPCR was performed using TB Green® Premix Ex Taq™ II kit (RR820A, Takara, Japan) according to the manufacturer’s instructions. The sequences of the amplification primers are shown in Table 1. The reaction was run on the 7500 Fast Real-time PCR system (Applied Biosystems). Gene expression was calculated by the 2−ΔCt × 1000 method. GAPDH was used as an internal control.
Flow cytometry
For surface staining, the PBMCs were incubated with anti-human CD4 monoclonal antibody (mAb) (#317407, BioLegend) for 20 min at room temperature. For intracellular staining, cells were fixed and permeabilized with a Fixation Buffer (#420801, BioLegend) and Intracellular Staining Perm Wash buffer (#421002, BioLegend), followed by intracellular staining with anti-human IFN-γ, IL-4, and IL-17 A mAb (#502531, 500808, 512333; BioLegend). All flow cytometry experiments were analyzed using a Beckman Coulter Fortessa cytometer.
Western blotting
Whole cell lysates were obtained from separated cells and cultured cells. Whole cell lysates were heated for 10 min at 100 °C in 5 × sodium dodecyl sulfate (SDS) loading buffer, separated by SDS-polyacrylamide gel electrophoresis, and transferred to polyvinylidene fluoride membranes. The membranes were blocked for 1 h at room temperature with 5% bovine serum albumin in 0.05% Tween 20/Tris buffered saline, followed by overnight incubation at 4 °C with primary antibody. The blots were incubated subsequently with secondary goat anti-rabbit horseradish peroxidase H-conjugated immunoglobulin G (IgG) for 1 h at room temperature. The reaction was visualized by chemiluminescence detection. The following primary antibodies were used: anti-Plexin-A1 (#M06377, Boster), anti-Plexin-B3 (#AF4958, R&D Systems), anti-PI3K (67071-1-Ig; Proteintech), anti-AKT (#4691, CST), anti-p-AKT (#4060, CST), anti-mTOR (#2972, CST), and anti-p-mTOR (#2971, CST).
Statistical analysis
GraphPad Prism v9.00 (GraphPad Software, San Diego, CA) was used for statistical analyses. For the normally distributed data, differences between groups were analyzed using Student’s t-test or paired t-test. For the non-parametric data, the differences between groups were analyzed by the Mann-Whitney U test or paired Wilcoxon tests. P < 0.05 was considered statistically significant, and all results were independently repeated at least 3 times.
Results
Elevated Sema5A positively correlated with Th17 polarization in SLE patients
To study the correlation of serum Sema5A with Th cytokines, Serum were collected from 33 SLE patients and 30 healthy controls. The clinical data of the samples were shown in Table S1. The levels of IFN-γ, IL-4, IL-17 A, IL-10 and soluble Sema5A were detected by ELISA. The results showed that serum levels of IL-17 A and IL-10 in SLE patients were significantly higher than those in healthy controls. Correlation analysis showed that serum Sema5A was positively correlated with IL-17 A levels in SLE patients (r = 0.5273, P = 0.0016), but no correlation with IFN-γ, IL-4, or IL-10 levels (Fig. 1). This suggests that the increased Sema5A might be involved in the Th17 polarization in SLE patients. As we have observed a connection between Sema5A and renal lesion of SLE in previous research, we further analyzed the correlation of serum Sema5A with Th cytokines in Lupus nephritis (LN) subgroup. Results showed that serum Sema5A were positively correlated with IL-17 A levels (n = 13, r = 0.5604, P = 0.0499), but had no statistical correlation with IFN-γ, IL-4, or IL-10 levels in LN patients (Figure S1).
Correlation between serum Sema5A and Th cytokine levels in SLE patients. A. Serum levels of IFN-γ, IL-4, IL-17 A, and IL-10 in SLE patients and healthy controls, (HC, n = 30 SLE, n = 33); B. Correlation between serum Sema5A and Th cytokine levels in SLE patients. Mann-Whitney U test, *: P < 0.05, **: P < 0.01, ****: P < 0.0001
Increased expression of PlexinA1 in CD4+ T cells from SLE patients
Since Sema5A strongly increased T cell and NK cell proliferation and induced the secretion of proinflammatory cytokines [19], we determined the expression of Sema5A receptors in SLE patients and healthy controls. The results showed that the mRNA level of PlexinA1 was higher in CD4+ T cells than that in CD8+ T cells, B cells and monocytes. Futhermore, compared with healthy controls, PlexinA1 levels were significantly increased in SLE patients. However, the PlexinB3 levels were extremely low, both in SLE patients and healthy controls. (Fig. 2A, B). Meanwhile, we analyzed a gene expression sequencing data set GSE164457 with 120 SLE patients in the GEO database and found that PlexinA1 level was significantly higher in CD4+ T cells than that in B cells, monocytes, and NK cells, while PlexinB3 could not be detected in most samples (Fig. 2C, D), which was consistent with our reports. We observed similar results from protein aspect using western blotting. (Fig. 2E-H). Furthermore, in another gene expression sequencing data set of Human CD4 + T cell subsets, GSE135390, we found the expression of PlexinA1 were mainly in Th subsets, especially Th17 and Th22, rather than in Naive CD4 + T cells or Regulatory T cells (Treg) (Figure S2).
PlexinA1 is elevated in CD4+T cells from SLE patients. A, B. The relative mRNA level of PlexinA1 and PlexinB3 in CD4+ T cells, CD8+ T cells, CD19+ B cells, and CD14+ monocytes from SLE patients and healthy controls (HC) a (n = 5–16); C, D. The mRNA level of PlexinA1 and PlexinB3 genes in CD4+ T cells, CD19+ B cells, CD14+ monocytes and NK cells from SLE patients in GEO dataset GSE164457 (n = 120); E. The protein level of PlexinA1 and PlexinB3 in CD4+ T cells, CD8+ T cells, CD19+ B cells, and CD14+ monocytes from SLE patients; F. The protein level of PlexinA1 and PlexinB3 in CD4+ T cells from SLE patients and healthy controls (HC); G. Semi-quantify protein level of PlexinA1 in CD4+ T, CD8+ T, CD19+ B, and CD14+ monocytes from SLE patients (n = 4); H. Semi-quantify protein level of PlexinA1 in CD4+ T from SLE patients and healthy controls (HC) (n = 6);.*: P < 0.05, **: P < 0.01, ***: P < 0.001, **** : P < 0.0001; ### / #### :P < 0.001 / P < 0.0001 compared to SLE CD4+ T cells, $$$ / $$$$ :P < 0.001 / P < 0.0001 compared to HC CD4+ T cells
Sema5A enhances Th17 polarization
Based on the results of previous studies, we further speculated that elevated Sema5A in SLE patients may promote Th17 polarization. Thus, PBMCs from SLE patients and healthy controls were activated with anti CD3/CD28 in the absence or presence of recombinant human Sema5A. In both SLE patients and healthy controls, Sema5A enhanced IL-17 A secretion induced by CD3/CD28 stimulation, as well as the expression of corresponding specific transcription factor RORC. In contrast, Sema5A did not regulate the expression of Th1, Th2 and Treg cytokines (Fig. 3).
Sema5A induced IL-17 A production in PBMCs from SLE patients and Healthy Controls. A. Expression of CD4+ T cells subset transcription factors in PBMCs from SLE patients and healthy controls; B. Th cytokines production by PBMCs from SLE patients and healthy controls; Cells were activated with anti-CD3/CD28 antibody in the absence or presence of Sema5A for 48 h. TBX21: T-Box Transcription Factor 21; GATA3: GATA Binding Protein 3; RORC: RAR Related Orphan Receptor C. Samples from the same donor were matched between the experimental group and the control group, a total of 6–8 cases. Wilcoxon paired rank sum test, *: P < 0.05, **: P < 0.01; Mann-Whitney U test, ##: P < 0.01
We next determined the proportion of CD4+ T cell subsets (Th1, Th2, and Th17) and CD4 negative cells from PBMCs by flow cytometry. The results showed that compared with control group, the proportion of CD4+IL-17 A+ T cells but not CD4+ IFN-γ+ or CD4+ IL-4+ cells was increased in the Sema5A stimulation group. There was no significant difference in the proportion of CD4− IFN-γ+, CD4− IL-4+, and CD4− IL-17 A+ cells between the Sema5A stimulation and control groups (Fig. 4). The above results suggest that Sema5A can promote the secretion of IL-17 A and increase the proportion of Th17 cells in PBMCs from SLE patients and healthy controls.
Sema5A promotes Th17 differentiation through PlexinA1
In order to further determine if Sema5A promotes CD4+T cells differentiation into Th17 cells directly, we separated CD4+ T cells from PBMCs and cocultured with or without Sema5A under Th0 and Th17 conditions. The results suggested that HC and SLE-derived CD4+ T cells secreted more IL-17 A under both Th0 condition and Th17 condition. Under Th0 condition, there was no significant difference in HC-derived CD4+ T cells between the Sema5A stimulation and PBS control, while the level of IL-17 A secreted by SLE-derived CD4+ T cells stimulated by Sema5A was higher than PBS control. Under Th17 condition, the levels of IL-17 A secreted by HC and SLE-derived CD4+ T cells after Sema5A stimulation were significantly higher than those in the PBS control (Fig. 5A). This suggested that Sema5A can promote the differentiation of HC and SLE CD4+ T cells into Th17 during in vitro differentiation. Next, we knocked down the expression of Receptor PlexinA1 in CD4+ T cells using siRNA (Fig. 5B) and cocultured with Sema5A. IL-17 A secretion in PlexinA1 knocked down CD4+ T cells was lower than NC after stimulation by Sema5A (Fig. 5C). This suggests that Sema5A promotes CD4+ T cells differentiation into Th17 cells through PlexinA1.
Sema5A promotes Th17 differentiation via PlexinA1. A. CD4+ T cells from HC and SLE patients were induced by Th0 cocktail (anti-CD3/CD28 antibody) or Th17 cocktail (anti-CD3/CD28 antibody, IL-1β, IL-6, IL-23, TGF-β1, anti-IFN-γ Antibody, anti-IL-4 antibody co-stimulation), Sema5A or PBS solution was added and cultured for 5 days, the levels of IL-17 A in the culture supernatant were detected by ELISA (n = 6 for HC and n = 8 for SLE patients, Cells from the same donor were paired between different groups). Wilcoxon test, *: P < 0.05, **: P < 0.01; Mann-Whitney U test,: P < 0.05. B. CD4+ T cells were activated by anti-CD3/CD28 antibody for 48 h in vitro, and then transfected with universal control siRNA (NC) or siRNA-Plexin A1s, protein expression of PlexinA1 was detected by Western blot. C. CD4+ T cells transfected with NC or siRNA-Plexin A1 were cultured with Sema5A or PBS solution for 5 days under Th17-inducing conditions, and the IL-17 A level in the culture supernatant was detected by ELISA. Tukey test *: P < 0.05, **: P < 0.01
Sema5A-PlexinA1 promotes Th17 differentiation via the activation of PI3K-Akt-mTOR pathway
We further detected the activation of the AKT-mTOR pathway in CD4+ T cells after induced with Sema5A. Results showed that Sema5A induction enhanced the phosphorylation of AKT(Ser473) and mTOR(Tyr2448) in Th17 cocktail stimulation for 1 h (Fig. 6A). Besides, this effect was reversed by inhibiting PlexinA1 using siRNA (Fig. 6B). Furthermore, we conducted rescue experiments using small molecule inhibitors of the PI3K/AKT/mTOR signaling pathway. When we added pictilisib (PI3K inhibitor), MK-2206 (AKT inhibitor), and temsirolimus (mTOR inhibitor) to the medium, the stimulatory effect of Sema5A on IL-17 A production was significantly abolished.
Sema5A promotes the activation of AKT-mTOR pathway in CD4 + T cells through PlexinA1. A. Activated CD4+ T cells were cultured for 24 h in medium containing Sema5A or PBS solution, followed by stimulating for 1 h with the Th17 cocktail; the phosphorylation of AKT and mTOR was detected by Western-blotting. B. Activated CD4+ T cells were transfected with siRNA-Plexin A1- or universal control siRNA (NC) for 24 h, and resuspended in fresh medium containing Sema5A or PBS solution for another 24 h, followed by stimulating for 1 h with the Th17 cocktail; the phosphorylation of AKT and mTOR was detected by Western-blotting. * : P < 0.05, ** : P < 0.01. C. Activated CD4+ T cells were cultured for 4 h in medium containing pictilisib (PI3K inhibitor), MK-2206 (AKT inhibitor), and temsirolimus (mTOR inhibitor), then cells were resuspended in medium with Sema5A or PBS solution for 24 h under Th17-inducing conditions, and the IL-17 A level in the culture supernatant was detected by ELISA. Tukey test *: P < 0.05
Discussion
We previously reported that soluble Sema5A elevated in SLE patients and correlated with disease activity and was involved in kidney and blood system damage [17]. However, the precise role of Sema5A in the development of SLE remains unknown. Here, in this study, a significantly positive correlation was identified between Sema5A expression and the production of IL-17 A in the serum of SLE patients. And we found that PlexinA1 in CD4+T cells as the most important receptor of Sema5A. Sema5A-PlexinA1 axis induced the production of IL-17 A by CD4+T cells. Taken together, these results demonstrate that Sema5A plays an essential role in Th17 differentiation in SLE.
As one of the “immune semaphorins”, Sema5A is involved in driving both innate and adaptive immune responses [19, 20], and it has been shown associated with the development of autoimmune diseases [14, 15, 17, 21]. It was reported that Sema5A greatly promoted T cell and natural killer (NK) cell proliferation and induced secretion of Th1/Th17 proinflammatory cytokines in RA [19]. Increased co-expression of Sema5A and IL-17 A in the skin of chronic spontaneous urticaria (CSU) also showed the relationship of Sema5A with IL-17 A [16]. Recently, Yang et.al reported anti-Sema5A treatment effectively improved the Treg/Th17 imbalance in collagen induced arthritis (CIA) mice, which indicated that Sema5A was involved in Th17 differentiation in RA [22]. In our study, we proved that Sema5A could significantly increase Th17 (CD4+IL-17 A+) cells as well as IL-17 A production, but did not regulate Th1, Th2 and Treg cells. In addition to Th17 cells, it has been reported that γδT cells, CD8+ T cells, and double negative T cells (DNT, CD3+ CD4−CD8−T) also secrete IL-17 A [7]. However, Sema5A treatment could not affect CD4−IL-17 A+ cells. This suggests that Sema5A may mainly promote IL-17 A production in CD4+T cells.
PlexinA1 and PlexinB3 are two major receptors of Sema5A, but the expression characteristics of these two receptors in immune cells remain unclear. We firstly found that PlexinA1 is mainly expressed in CD4+T cells but not B cells, monocytes, or NK cells. While PlexinB3 can rarely be detected in PBMCs. Bulk RNA sequencing data from GEO database confirmed this point. We further demonstrated that Sema5A promotes CD4+T cells differentiation into Th17 cells via connecting with PlexinA1, blocking PlexinA1 disrupts IL-17 A production.
Previous studies have reported that Th17/IL-17 A plays a critical role in the pathogenesis of LN [3, 23]. Immune complexes and other inflammatory factors stimulate renal cells producing chemokines such as CCL20, CXCL9, and CXCL10, thus recruiting Th17 cells into renal tissue to mediate tissue inflammation [24,25,26,27]. Th17/IL-17 A in renal tissue can change the function and structure of renal tissue cells, and promote the inflammatory environment [28, 29]. IL-17 A can also induce the expression of chemokines in renal tissue cells attracting inflammatory cells, especially neutrophils to infiltrate and further aggravate tissue damage and prevent tissue repair, ultimately leading to renal fibrosis and loss of renal function [23, 30, 31]. In addition to its role in LN, a recent study shows that IL-17 A can promote the survival of long-lived plasma cells in lupus mice and increase the secretion of pathogenic autoantibodies. These plasma cells cannot be eliminated by traditional targeted B cell therapy, so targeting IL-17 A may become a therapeutic target for some refractory lupus [32, 33].
The PI3K-Akt-mTOR pathway is one of the most important intracellular pathways involved in cell proliferation, growth, differentiation and survival. mTOR has recently emerged as a key regulator of T cell proliferation and differentiation [34]. mTOR complex 1 is essential for Th1 and Th17 differentiation [35, 36]. Previous mouse studies have investigated that mTORC1 inhibitor rapamycin inhibits glomerulonephritis in lupus-prone mice by reducing Th17 cells and promoting Treg cell function [37, 38]. Clinically, the treatment of targeting T cells by inhibiting mTORC1 through rapamycin has played a significant role in the treatment of connective tissue-associated thrombocytopenia [39].Recent studies have shown that rapamycin can reverse the proliferation of Th17 cells in SLE patients and may become a target for SLE treatment [40, 41]. Interestingly, in the following mechanism study, we found that Sema5A-PlexinA1 axis activated PI3K/Akt/mTOR pathway in Th17 differentiation. Therefore, inhibiting Sema5A to reduce mTOR activation in CD4+ T cells and limit Th17 differentiation may also be a potential therapeutic target for SLE.
In addition to IL-17 A, IFN-γplays an important role in pathogenesis of LN as well [42]. In our study, although not statistically significant, we found that there is a tendency that serum Sem5A might be positively correlated with serum IFN-γ levels and serum IFN-γto IL-4 ratios (Figure S1). Considering the heterogeneity of SLE and limited serum samples, the variation of Sema5A levels was not small. Therefore, we analyzed the Top 5 serum Sema5A level cases of the results. It showed that these cases have higher serum IgG, lower complement C4, and higher SLEDAI-2000 disease activity sores (Table S2). Thus, large cohort of SLE /LN patients is required in the future to identify the connection between Sema5A and IFN-γ, and other inflammatory cytokines.
Although neutralizing Sema5A in CIA model significantly reduced mice serum IL-17 A levels [43], the promotion effect of Sema5A in IL-17 A release by CD4+T cells in vitro seems weak in our study, which indicates other indirect mechanisms may exist in vivo to promote IL-17 A/Th17. Intriguingly, our study in RA has shown that Sema5A can promote the release of other inflammatory cytokines including IL-6 in fibroblast [21]. And IL-6 is an important mediator for Th17 differentiation. Therefore, future studies are required to clarify more in vivo mechanisms of Sema5A in SLE pathogenesis.
In conclusion, our findings suggest that Sema5A plays an important role in SLE pathology, through the induction of IL-17 A by CD4+T cells. Its effect may be achieved by activating the PI3K-Akt-mTOR pathway via the PlexinA1 receptor in CD4+ T cells. This study preliminarily provides a basis for Sema5A as a potential target for SLE treatment.
Data availability
No datasets were generated or analyzed during the current study.
References
Kaul A, Gordon C, Crow MK, Touma Z, Urowitz MB, van Vollenhoven R, Ruiz-Irastorza G, Hughes G. Systemic Lupus Erythematosus. Nat Rev Dis Primers. 2016;2(1):16039. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrdp.2016.39.
Kiriakidou M, Ching CL. Systemic Lupus Erythematosus. Ann Intern Med. 2020;172(11):ITC81–96. https://doiorg.publicaciones.saludcastillayleon.es/10.7326/AITC202006020.
Aw YTV, Whiley PJ, Lorenzo AM, Lea-Henry T, Shanmuganandam S, Stanley M, Babu SN, Athanasopoulos V, Cappello J, Ellyard JI, et al. Immunophenotyping identifies distinct Cellular signatures for systemic Lupus Erythematosus and Lupus Nephritis. Rheumatol Autoimmun. 2023;3(1):15–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/rai2.12060.
Shi X, Wang Z, Tang R, Chen WP. Injury and Death: New insights into Lupus Nephritis Pathogenesis and Therapy. Rheumatol Autoimmun. 2023;3(4):193–204. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/rai2.12103.
Zhu JT. Helper cell differentiation, heterogeneity, and plasticity. Cold Spring Harb Perspect Biol. 2018;10(10):a030338. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a030338.
Cheng Q, Chen X, Xu J, Chen M, Zhang P, Wu H, Du Y. lncRNA X-inactive‐specific transcript is a potential biomarker related to changes in CD4 + T cell levels in systemic Lupus Erythematosus. Rheumatol Autoimmun. 2022;2(3):159–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/rai2.12048.
Crispín JC, Oukka M, Bayliss G, Cohen RA, Van Beek CA, Stillman IE, Kyttaris VC, Juang Y-T, Tsokos GC. Expanded Double Negative T Cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol. 2008;181(12):8761–6. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.181.12.8761.
Garcia S. Role of semaphorins in Immunopathologies and Rheumatic diseases. IJMS. 2019;20(2):374. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms20020374.
Nishide M, Kumanogoh A. The role of Semaphorins in Immune responses and Autoimmune Rheumatic diseases. Nat Rev Rheumatol. 2018;14(1):19–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2017.201.
Koda T, Namba A, Kinoshita M, Nakatsuji Y, Sugimoto T, Sakakibara K, Tada S, Shimizu M, Yamashita K, Takata K, et al. Sema4A is implicated in the acceleration of Th17 cell-mediated neuroinflammation in the Effector Phase. J Neuroinflammation. 2020;17(1):82. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-020-01757-w.
Goldberg JL, Vargas ME, Wang JT, Mandemakers W, Oster SF, Sretavan DW, Barres BA. An Oligodendrocyte Lineage-Specific Semaphorin, Sema5A, inhibits Axon Growth by Retinal Ganglion cells. J Neurosci. 2004;24(21):4989–99. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.4390-03.2004.
Artigiani S, Conrotto P, Fazzari P, Gilestro GF, Barberis D, Giordano S, Comoglio PM, Tamagnone L. Plexin-B3 is a functional receptor for semaphorin 5A. EMBO Rep. 2004;5(7):710–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.embor.7400189.
Matsuoka RL, Chivatakarn O, Badea TC, Samuels IS, Cahill H, Katayama K-I, Kumar SR, Suto F, Chédotal A, Peachey NS, et al. Class 5 transmembrane Semaphorins Control Selective mammalian retinal lamination and function. Neuron. 2011;71(3):460–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuron.2011.06.009.
Lyu M, Li Y, Hao Y, Sun T, Liu W, Lyu C, Fu R, Li H, Xue F, Liu X, et al. Elevated semaphorin 5A correlated with Th1 polarization in patients with chronic Immune Thrombocytopenia. Thromb Res. 2015;136(5):859–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.thromres.2015.07.032.
Liu Y, Wang S, Guo Q, Li Y, Qin J, Zhao N, Li Y, Shan Z, Teng W. Elevated semaphorin 5A in patients with Hashimoto’s Thyroiditis: a case-control study. Endocr Connect. 2017;6(8):659–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1530/EC-17-0132.
Lobna M, Nasren E, Adi S, Jacob B, Maria G-AA, Marcus M, Zahava V. The expression of IL-17, in chronic spontaneous urticaria is linked to Semaphorin5A. Biomolecules. 2021;11(3):373. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom11030373.
Du Y, Wu X, Chen M, Wang W, Xv W, Ye L, Wu D, Xue J, Sun W, Luo J, et al. Elevated semaphorin5A in systemic Lupus Erythematosus is in Association with Disease Activity and Lupus Nephritis. Clin Exp Immunol. 2017;188(2):234–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cei.12924.
Hochberg MC. Updating the American College of Rheumatology Revised Criteria for the classification of systemic Lupus Erythematosus. Arthritis Rheum. 1997;40(9):1725. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.1780400928.
Gras C, Eiz-Vesper B, Jaimes Y, Immenschuh S, Jacobs R, Witte T, Blasczyk R, Figueiredo C. Secreted semaphorin 5A activates Immune Effector cells and is a biomarker for rheumatoid arthritis: secreted semaphorin 5A in rheumatoid arthritis. Arthritis Rheumatol. 2014;66(6):1461–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.38425.
Chen M-Z, Su L-Y, Ko P-H, Hsu M-H, Chuang L-L, Chen L-H, Lu T-P, Chuang EY, Chow L-P, Tsai M-H, et al. Extracellular domain of semaphorin 5A serves a tumor–suppressing role by activating Interferon Signaling pathways in Lung Adenocarcinoma cells. Int J Oncol. 2022;60(2):21. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/ijo.2022.5311.
Cheng Q, Chen M, Liu M, Chen X, Zhu L, Xu J, Xue J, Wu H, Du Y. Semaphorin 5A suppresses ferroptosis through activation of PI3K-AKT-mTOR signaling in rheumatoid arthritis. Cell Death Dis. 2022;13(7):608. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-022-05065-4.
Qin Y, Jin J, Zhang J, Wang H, Liu L, Zhang Y, Ling S, Hu J, Li N, Wang J, et al. A fully human monoclonal antibody targeting semaphorin 5A alleviates the progression of rheumatoid arthritis. Biomed Pharmacother. 2023;168:115666. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2023.115666.
Paquissi FC, Abensur H. The Th17/IL-17 Axis and kidney diseases, with Focus on Lupus Nephritis. Front Med (Lausanne). 2021;8:654912. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmed.2021.654912.
Krohn S, Nies JF, Kapffer S, Schmidt T, Riedel J-H, Kaffke A, Peters A, Borchers A, Steinmetz OM, Krebs CF, et al. IL-17 C/IL-17 receptor E signaling in CD4 + T cells promotes TH17 cell-driven glomerular inflammation. J Am Soc Nephrol. 2018;29(4):1210–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1681/ASN.2017090949.
Petrovic-Djergovic D, Popovic M, Chittiprol S, Cortado H, Ransom RF, Partida-Sánchez S. CXCL10 induces the recruitment of monocyte-derived macrophages into kidney, which aggravate Puromycin Aminonucleoside Nephrosis. Clin Exp Immunol. 2015;180(2):305–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cei.12579.
Panzer U, Steinmetz OM, Paust H-J, Meyer-Schwesinger C, Peters A, Turner J-E, Zahner G, Heymann F, Kurts C, Hopfer H, et al. Chemokine receptor CXCR3 Mediates T Cell Recruitment and Tissue Injury in Nephrotoxic Nephritis in mice. J Am Soc Nephrol. 2007;18(7):2071–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1681/ASN.2006111237.
Lu G, Zhang X, Shen L, Qiao Q, Li Y, Sun J, Zhang J. CCL20 secreted from IgA1-Stimulated Human Mesangial Cells Recruits Inflammatory Th17 Cells in IgA Nephropathy. PLoS ONE. 2017;12(5):e0178352. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0178352.
Yan J, Li Y, Yang H, Zhang L, Yang B, Wang M, Li Q. Interleukin-17A participates in Podocyte Injury by inducing IL-1β secretion through ROS-NLRP3 inflammasome-Caspase-1 pathway. Scand J Immunol. 2018;87(4):e12645. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/sji.12645.
Zhai S, Sun B, Zhang Y, Zhao L, Zhang L. IL-17 aggravates renal Injury by promoting Podocyte Injury in Children with primary nephrotic syndrome. Exp Ther Med. 2020;20(1):409–17. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/etm.2020.8698.
L L, Fg L, M Y, L W, Y C, L W, W J, Jm F. Effect of pro-inflammatory Interleukin-17A on epithelial cell phenotype inversion in HK-2 cells in Vitro. Eur Cytokine Netw. 2016;27(2). https://doiorg.publicaciones.saludcastillayleon.es/10.1684/ecn.2016.0373.
Zhang Y, Chen X, Deng Y. miR-125a-3p decreases levels of Interlukin-17 and suppresses renal fibrosis via down-regulating TGF-Β1 in systemic lupus erythematosus mediated Lupus Nephritic mice. Am J Transl Res. 2019;11(3):1843–53.
Ma K, Du W, Xiao F, Han M, Huang E, Peng N, Tang Y, Deng C, Liu L, Chen Y, et al. IL-17 sustains the plasma cell response via P38-Mediated Bcl-xL RNA Stability in Lupus Pathogenesis. Cell Mol Immunol. 2021;18(7):1739–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41423-020-00540-4.
Robert M, Miossec P. Interleukin-17 and Lupus: enough to be a target? For which patients? Lupus. 2020;29(1):6–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0961203319891243.
S J, J H, X Y, T. Cell metabolism: a New Perspective on Th17/Treg Cell Imbalance in systemic Lupus Erythematosus. Front Immunol. 2020;11. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2020.01027.
Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD. The kinase mTOR regulates the Differentiation of Helper T Cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol. 2011;12(4):295–303. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.2005.
Li H, Boulougoura A, Endo Y, Tsokos GC. Abnormalities of T cells in systemic Lupus Erythematosus: New insights in Pathogenesis and therapeutic strategies. J Autoimmun. 2022;102870. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaut.2022.102870.
Zhang J, Guo Q, Dai D, Yu J, Wang L, Wu Z, Ding H, Shen N, Duan Y. Rapamycin-Encapsulated Costimulatory ICOS/CD40L-Bispecific nanoparticles restrict pathogenic helper T-B-Cell interactions while in situ suppressing mTOR for Lupus Treatment. Biomaterials. 2022;289:121766. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biomaterials.2022.121766.
Song X, Gao J, Liu H, Liu X, Tang K. Rapamycin alleviates renal damage in mice with systemic lupus erythematosus through improving Immune response and function. Biomed Pharmacother. 2021;137:111289. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2021.111289.
Wu C, Wang Q, Xu D, Li M, Zeng X. Sirolimus for patients with connective tissue disease-related refractory thrombocytopenia: a Single-Arm, open-label clinical trial. Rheumatology (Oxford). 2021;60(6):2629–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keaa645.
Kato H, Perl A. Mechanistic Target of Rapamycin Complex 1 expands Th17 and IL-4 + CD4-CD8- Double-Negative T Cells and Contracts Regulatory T Cells in systemic Lupus Erythematosus. J Immunol. 2014;192(9):4134–44. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1301859.
Lai Z-W, Borsuk R, Shadakshari A, Yu J, Dawood M, Garcia R, Francis L, Tily H, Bartos A, Faraone SV, et al. Mechanistic target of Rapamycin activation triggers IL-4 production and necrotic death of double-negative T cells in patients with systemic Lupus Erythematosus. J Immunol. 2013;191(5):2236–46. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1301005.
Jacob CO, van der Meide PH, McDevitt HO. Vivo treatment of (NZB X NZW)F1 lupus-like nephritis with monoclonal antibody to Gamma Interferon. J Exp Med. 1987;166(3):798–803. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.166.3.798.
Xiao C, Lv C, Sun S, Zhao H, Ling H, Li M, Qin Y, Zhang J, Wang J, Yang X. TSP1 is the essential domain of SEMA5A involved in Pannus formation in rheumatoid arthritis. Rheumatology. 2021;keab133. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keab133.
Funding
The Traditional Chinese Medicine Science and Technology Project of Zhejiang Province (No.2024ZL099) and National Natural Science Foundation of China (No. 82271817) funded this study. The Chinese National Key Technology R&D Program (2021YFC2501300).
Author information
Authors and Affiliations
Contributions
YD, and XC contributed to the research design and drafted the manuscript. XC, and LJZ performed the in vitro experiments, XC, LJZ, and YJD collected clinical samples and clinical data, QC, and JYX assisted in experiments design and manuscript writing. JYX, YFX, and LH contributed to manuscript revision. All authors approved the submitted version of the manuscript and agreed to accept responsibility for all its aspects.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The Ethics Committee of the Second Affiliated Hospital of Zhejiang University School of Medicine approved this study (the approval protocol No: 2020433). Before enrollment, all patients gave their written informed consent.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Chen, X., Zhu, L., Xu, J. et al. Semaphorin 5A promotes Th17 differentiation via PI3K-Akt-mTOR in systemic lupus erythematosus. Arthritis Res Ther 26, 204 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-024-03437-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-024-03437-z