Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease with multiple etiological factors. Immune disorder contributes to SLE development and is an important clinical manifestation of SLE patients. Immune dysfunction is characterized by abnormal of B cells, T cells, monocyte-macrophages and dendritic cells (DCs), in both quantity and quality. Adenosine is a critical factor for human immune homeostasis, which acts as an immunosuppressive signal and can prevent the hyperactivity of human immune system. Adenosine levels are significant decreased in serum from SLE patients. Adenosine level is regulated by the CD39, CD73 and adenosine deaminase (ADA). CD39/CD73/ADA catalyzed the cascade enzymatic reaction, which contained the adenosine generation and degradation. Adenosine affects the function of various immune cells via bind to the adenosine receptors, which are expressed on the cell surface. This review aims to export the changes of immune cells and adenosine signal pathway in SLE, as well as the effect of adenosine signal pathway in SLE development.
Keywords: CD39, CD73, Adenosine deaminase, Adenosine, Systemic lupus erythematosus
1. Introduction
Systemic lupus erythematosus (SLE) is a multisystem inflammatory autoimmune disease, which is usually affects young and middle-aged women, characterized by production of autoantibodies and autoimmune inflammation. There are multiple factors involved in SLE pathogenesis and development, such as genetics, infection, hormone disorder and immune anomalies [1,2]. Among these factors, immune dysfunction has been found to be the critical driving factor for SLE, which characterized by loss of immune tolerance and overactive immune response. The dysfunction of B cell, T cell, macrophage and dendritic cell (DCs) were all involved in SLE development [[3], [4], [5]].
Adenosine plays as an immunosuppressive signal and shields tissues from an excessive inflammatory response and immune-mediated damage [6]. Zhang et al.’s study has showed that adenosine level was significant decreased in serum from SLE patients [7]. Adenosine metabolism is constituted by enzymes cascade as followed: ATP/ADP were degraded into AMP, which catalyzed by CD39 (ecto-nucleoside triphosphate diphosphohydrolase 1, E-NTPDase1); AMP was degraded into adenosine, which catalyzed by CD73 (ecto-5′-nucleotidase, NT5E); adenosine was degraded into inosine, which catalyzed by ADA. Thus, the dynamic equilibrium of adenosine concentration in vivo were regulated directly by the activities of CD39/CD73/ADA [6]. Human ADA contains two isoenzymes: ADA1 and ADA2. ADA1 is a predomination intracellular enzyme, and play important roles in human immune system development [8,9]. ADA1 gene defects would cause the severe combined immunodeficiency (SCID). ADA2 is a secreted protein produced by monocyte/macrophage [10]. Notably, beside as a adenosine deaminase, ADA2 also belonged to adenosine deaminase growth factor (ADGF) family [11], and could bind to certain immune cell subtype via proteoglycans or other unknown receptor [12]. In our previous study, we have found that ADA activity was significant increased in SLE patients and correlated with the SLE disease activity [13].
There were four types of adenosine receptor (i.e. A1R, A2AR, A2BR and A3R), which were expressed on cytomembrane of various immune cells. Via binding to these adenosine receptors, adenosine regulated a wide variety of cellular processes, including cell development and differentiation, cytokines secretion, cell activity and etc, which behaving as inflammatory response down-regulation and anti-inflammatory immunity up-regulation [14]. Thus, adenosine signal pathway is important for the immune homeostasis and is involved in the pathogenesis of autoimmune diseases.
This study aimed to export the changes of immune cells and adenosine signal pathway in SLE, as well as the effect of adenosine signal pathway in SLE development. In this paper, “adenosine” and “systemic lupus erythematosus” were used as key words to search articles (2002–2022) in PubMed database. Searches were conducted by the authors on Jun 18, 2022. 149 total databases were searched. Correspondence, comment and letters were excluded. Then, manual searches for the reference lists of the literatures was conducted to find potential eligible studies. Finally, 79 papers were included in this study.
Several limitations of this scoping review should be acknowledged. Although we adopted a comprehensive search, relevant studies may have been missed. The present review did not include literature written in languages other than English. This review did not assess risk of bias.
2. Adenosine pathway regulated the immune cells of SLE
2.1. Adenosine pathway in the regulation of B cells
The production of autoantibodies resulting from the breach of B cell tolerance in SLE patients, which demonstrated the critical role of B cells during the SLE progress. The BAFF (B cell activating factor), also known as Blys (B lymphocyte stimulator), is important in activation, differentiation and survival of B cells. Excess BAFF supports self-reactive B cells and prevent their deletion [15]. Several studies have showed the elevated levels of soluble Blys (sBlys) in serum from SLE patients [16,17]. Belimumab, a recombinant immunoglobulin G1λ human monoclonal antibody targeting Blys, has been approved for the treatment of active SLE [18]. These data demonstrated the important role of B cells abnormality in SLE development.
B cells expressed CD39, CD73 and adenosine receptors. Saze Z showed that 2-chloroadenosine (a adenosine analogue) inhibited B cells proliferation and cytokines expression, and A3R selective antagonist restored B cells functions, which presenting an important role of adenosine signal in B cell biology [19]. Hesse J and colleagues found that the enzymatic activity of CD73 in SLE B cells was almost fully abolished, despite unaltered expression of CD73. while the expression and enzymatic activity of CD39 were not changed [20]. The CD73 inactivation in B cells might contribute to SLE development, because of the less adenosine production. Consistently, CD73 deletion in mice leaded to higher levels of auto-antibodies in response to pristane, and promoted expansion of activated B cells and plasma cells [21]. Dang J and colleagues found that Human gingiva derived MSCs (GMSCs) could alleviate lupus nephritis histopathological scores by suppressing B cells activation, proliferation and differentiation [22]. Notably, the suppressive capacities of GMSCs could be abrogated by CD39−CD73 pathway blockage. CD11c+ T-bet+ B cells were recognized as an important component of humoral immunity and autoimmunity. A2AR agonists could reduce anti-nuclear antibodies and kidney pathology in lupus-prone mice by depleting CD11c + T-bet + B cells [23]. Correspondingly, Bortoluzzi A found that A2AR were up-regulated in lymphocytes from SLE patients (vs healthy subjects). Moreover, there was a negative correlation between the A2AR expression levels and SLE disease activity [24].
2.2. T cells
2.2.1. T cells in SLE
T cells play important roles in human immune tolerance. Several T cell subsets abnormalities have been described in SLE patients, including regulatory T cells (Treg) and Helper T cell (Th). Treg contributes to immune homeostasis and tolerance. Treg deficits, both quantitative and functional, have been found to be a trigger for SLE development. As shown in Fig. 1, the CD4+CD25+Foxp3+ Treg percentage in CD4+ T cells was significant lower in SLE patients compared to healthy controls [[25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37]]. Moreover, Kamal M's study showed that Treg percentage negatively correlated to SLE disease activity [38]. Valencia X found that Treg from patients with active SLE showed poorly immunosuppressive activity [39]. Notably, Treg/Th17 imbalance have been found to be involved in SLE development. Th17/Treg ratio was positively correlated with SLE disease activity [40]. Via restoring the number of Treg and the balance of Th17/Treg cells, SLE disease activity could be remission [[41], [42], [43]].
Fig. 1.
Forest plot showed the Treg cells percentage in SLE patients. 13 studies were used for the forest plot analysis. The total number of healthy control and SLE patients were 334 and 465 respectively.
2.2.2. Adenosine pathway in regulating of T cells
Accumulated studies have demonstrated the roles of CD39 and CD73 for the immune suppressive activity of Treg cells. Smyth LA's study found that CD73 present on Treg cell derived exosomes was essential for their suppressive function. The suppressive capacity was lacking in the
exosomes derived from a CD73-negative Treg cells [44]. Expanded human Treg in vitro show enhanced suppressive function, which was associated with stable CD73 expression. Co-expression of CD39/CD73 enabled expanded Tregs to convert ATP to adenosine, which is important for its suppressive activity [45]. Moreover, CD39 and CD73 played important roles in regulating T cell differentiation. CD39 and CD73 expression were finely regulated during the transition of naive/memory T cells to effector cells. Naive/memory T cells were endowed with the ability to produce adenosine and lose this ability as they transition to more differentiated cells [46].
ADA also played important roles in T cells immunity, including precursor T cell differentiation, Treg function and Follicular helper T cells (Tfh) function [[47], [48], [49]]. Naval-Macabuhay I's study showed that ADA could lead to a decrease of Treg cells and enhancement of CD4+ T cells, which would revert Treg cell-mediated inhibition of immune responses and restore T lymphocyte function in HIV-1 infection [50]. Tardif V et al.’s study have showed that ADA1 expression and activity controled the Tfh program by influencing IL6/IL-2 production [51]. Wu Z's study showed that, although there was no significant alteration of T cell subsets in patients with ADA2 deficiency (DADA2), T cells in DADA2 patients showed distinct cell-cell interactions with monocytes [52].
Adenosine receptors are important for T cell immunity. Ohta A's study showed that Treg cells cultured with A2AR agonist showed increased expression of CTLA-4 and stronger immunosuppressive activity. There was a significant increase of Treg cell number after A2AR stimulation. A2AR-deficiency in Tregs reduces their immunosuppressive efficacy in vivo [53,54]. A2BR was involved in the induction of Tregs. Treg induction could be promoted by A2BR agonist and prevented by A2BR-deficiency [55].
2.3. Monocyte/macrophage
2.3.1. Monocyte/macrophage in SLE
Monocyte/macrophages is the important component of the human phagocytic system, which is critical for elimination of pathogens and apoptotic cells. Circulating monocytes can be recruited to tissues and differential into macrophages. Breakdown of immune tolerance in SLE, at least in part, may result from the accumulation of apoptotic cells, which releases a lot of auto-antigens and subsequent induces auto-immune response. Several studies have showed that the phagocytic ability of macrophages to apoptotic cells was decreased in SLE patients [[56], [57], [58]].
The imbalance of M1/M2 polarization of macrophages was closely related to SLE development. The increase of pro-inflammatory M1 cells were found in SLE, and positively correlated to the SLE disease activity [[59], [60], [61], [62]]. Adoptive transplantation M2 cells or inducing of M2 cell proliferation in vivo could alleviate SLE severity in mice models [63,64].
2.3.2. Adenosine pathway in regulating of monocyte/macrophage
Current studies showed that adenosine could promote macrophages polarization toward to M2 and inhibit M1 macrophage activity via binding adenosine receptors [65]. Zavialov AV's study showed that ADA2 could induce monocytes differentiation into macrophages, and stimulates proliferation of T helper cells and macrophages [66]. By using monocyte derived macrophages cells, Tiwari-Heckler S et al.'s study showed that ADA2 treatment could induce up-regulation of pro-inflammatory cytokines: TNFα, IL-1, IL-6 (M1 markers), and downregulation of CD163 (M2 marker) [67]. However, Zhu and colleagues showed that ADA2 promotes M2 polarization of microglia/macrophages [68]. Together, these evidences suggested that the effect of ADA2 on macrophage differentiation may be complex.
CD73 also found to be involved in macrophage polarization process. Xu S et al.'s study showed that CD73 overexpression could promote macrophages/microglia M2 polarization in mice [69]. Consistently, Costales MG et al.'s study showed that CD73 inhibited macrophage produced more NO and a higher levels of pro-inflammatory cytokines (IL-1β, TNF-α), which was a characteristics of M1 macrophages [70].
2.4. Adenosine pathway in the regulation of DCs
DCs present phagocytosed antigens to naive T cells and are essential for immunity. Studies have showed that DCs as essential players in the mechanisms underlying SLE. Myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) represent the two major DCs subsets in blood [71]. Chan VS and colleagues have summarized the distinct roles of mDCs and pDCs in SLE [72]. In summary, DCs abnormalities are often observed in SLE patients. Inappropriate activation of DCs leaded to a breach of tolerance and activation of autoreactive T- and B-lymphocytes. The studies have found that unabated activation of DCs through IFN-α produced by pDCs may drive the autoimmune response in SLE [73,74].
Differentiation of functional DCs depends on the microenvironment. The ATP-adenosine balance controls by CD39/CD73 play an important role in DCs regulation [75]. In hypoxic or inflamed tissues, adenosine reached high physiologically levels and regulated the differentiation and immune function of DCs through adenosine receptor. Novitskiy SV et al.’s study showed that adenosine activation skewed DCs differentiation toward a distinct cell population characterized by impaired allostimulatory activity and high expression of angiogenic, pro-inflammatory, immune suppressor, and tolerogenic factors [76]. Liu C et al.’s study showed that stimulation of adenosine-A2AR axis could inhibit the maturation and proinflammatory function of reoxygenated DCs [77]. Moreover, Silva-Vilches C and colleagues found that generation of adenosine by CD73+ DCs was a crucial mechanism for the induction of anergic T cells and tolerance [78]. In vitro, ADA1 induced mDC maturation and ADA1 treatment also promoted the secretion of the IL-6 from mDCs [79].
3. CD39/CD73/ADA1/ADA2 expression levels in immune cells from SLE
Due to the important roles of adenosine metabolism pathway in SLE, we analyzed the expression change of CD39, CD73, ADA1 and ADA2 in SLE peripheral blood mononuclear cell (PBMC) and immune cells subsets, including B cell, T cell and Monocyte based on the GEO database (Table 1), by searching GSE datasets about blood cells from SLE patients in GEO database. As shown in Fig. 2, compared with healthy controls, CD39 and ADA2 expression levels were significant increased in PBMC from SLE patients (Fig. 2A and B). Based on GSE4588 dataset, CD39 was decreased and ADA1 was increased in B cells from SLE, while based on GSE10325 dataset for B cells, there were no significant difference of the expression of CD39, CD73, ADA1 and ADA2 between SLE and healthy controls (Fig. 2C and D). In T cells (SLE vs healthy controls), ADA1 expression was increased base on GSE4588 dataset and CD73 was decreased based on GSE10325 dataset (Fig. 2E and F). In monocyte cells, GSE46904 dataset showed that there were no significant difference of CD39/CD73/ADA1/ADA2 expression between SLE and healthy controls (Fig. 2G). Taken together, these data showed the expression change of molecules related to adenosine metabolism, which indicated that adenosine metabolism might be involved in SLE development.
Table 1.
Summary of GEO datasets included in this study.
| GEO accession | Platforms | Patients | Controls | Cell type |
|---|---|---|---|---|
| GSE50772 | GPL570 | 61 SLE | 20 healthy | PBMC |
| GSE81622 | GPL10558 | 30 SLE | 25 healthy | PBMC |
| GSE4588 | GPL570 | 7 SLE | 9 healthy | CD19+ B cell |
| GSE10325 | GPL96 | 14 SLE | 9 healthy | CD19+ B cell |
| GSE4588 | GPL570 | 8 SLE | 10 healthy | CD4+ T cell |
| GSE10325 | GPL96 | 14 SLE | 9 healthy | CD4+ T cell |
| GSE46907 | GPL96 | 5 SLE | 5 healthy | CD14+ Monocyte |
Fig. 2.
The difference of expression levels for CD39, CD73, ADA1 and ADA2 in SLE and controls base on GEO database. Differential expression analysis of CD39, CD73, ADA1 and ADA2 in PBMC (A, B), B cells (C, D), T cells (E, F) and monocyte (G). Student's t-test was used to analyze the difference expression levels between controls and SLE. P < 0.05 was considered to be statistically significant. R software was used to perform statistical analysis and mapping.
4. Adenosine receptors expression in SLE PBMC and immune cells
Adenosine receptors were important for the immune regulated function of adenosine. Thus, we also analyzed the expression change of A1R, A2AR, A2BR and A3R in SLE PBMC and immune cells subsets. As shown in Fig. 3, A2BR expression was significant higher in SLE PBMC than that in healthy controls (Fig. 3A and B). Moreover, GSE50772 dataset showed A3R was significant increased in SLE PBMC (Fig. 3B). However, in B cells, T cells and monocytes, there were no significant difference of four adenosine receptors between SLE and healthy controls (p > 0.05, Fig. 3C–G). Taken together, these data indicated the expression of adenosine receptors might be not extensive changed in SLE patients. Thus, the function of adenosine-receptor signal pathway in SLE might be not regulating by the adenosine receptor expression.
Fig. 3.
The difference of expression levels for A1R, A2AR, A2BR and A3R in SLE and controls base on GEO database. Differential expression analysis of A1R, A2AR, A2BR and A3R in PBMC (A, B), B cells (C, D), T cells (E, F) and monocyte (G). Student's t-test was used to analyze the difference expression levels between controls and SLE. P < 0.05 was considered to be statistically significant. R software was used to perform statistical analysis and mapping.
5. Concluding remarks
SLE is a complex and systemic autoimmune disease with multiple etiological factors. A better understanding of immunopathogenic mechanisms involved in SLE may be helpful in exploring SLE origin, finding biomarker for diagnose and illness monitoring, and screening the potential treatment targets.
Adenosine is a key signal for human immunity homeostasis, which acts as a “alarm signal” and prevents an overactive immune response. The concentration of adenosine in human is depended by CD39/CD73/ADA1/ADA2, and the effects of adenosine on immune cells are correlated with the adenosine driven-adenosine receptors activation. In this review, we explored the change of immune cells in SLE, and the influence of adenosine signal related molecules on immune cells, including B cells, T cells, monocyte/macrophage and DCs. In summary, the change of quantity or quality for these immune cells are usually involved in SLE development. And the expression or activity levels of CD39/CD73/ADA1/ADA2 may be involved in regulating of these immune cells. Transcriptome sequencing datasets showed that the expression levels of CD39/CD73/ADA1/ADA2 were often changed in immune cells from SLE patients, while the expression of adenosine receptors were not changed (Fig. 2, Fig. 3). To date, the studies on adenosine pathway in SLE are still few, for a better understanding of the immune dysregulation in SLE, more studies involving the adenosine signal pathway in SLE are needed.
Funding
This work is supported by the Scientist Fund of Tangdu hospital (2021SHRC004; 2021LCYJ049 and 2021SHRC031).
Ethics declarations
Review and/or approval by an ethics committee was not needed for this study because this study is based exclusively on published literature. For the same reason, informed consent was not required.
Data availability statement
There is no research related data stored in publicly available repositories, and the data in this paper will be made available on request from the corresponding author.
CRediT authorship contribution statement
Ke Dong: Writing – original draft. Xia-nan Wu: Writing – original draft. Ying-qi Liu: Writing – review & editing. Lan Yang: Data curation. Chong Liu: Software. Hui-ping Wang: Investigation. Zhao-wei Gao: Writing – review & editing, Supervision.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- 1.Robinson G.A., Wilkinson M., Wincup C. The role of Immunometabolism in the pathogenesis of systemic lupus erythematosus. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.806560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tsokos G.C., Lo M.S., Costa R.P., Sullivan K.E. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat. Rev. Rheumatol. 2016;12:716–730. doi: 10.1038/nrrheum.2016.186. [DOI] [PubMed] [Google Scholar]
- 3.Szelinski F., Lino A.C., Dorner T. B cells in systemic lupus erythematosus. Curr. Opin. Rheumatol. 2022;34:125–132. doi: 10.1097/BOR.0000000000000865. [DOI] [PubMed] [Google Scholar]
- 4.Yuan S., Zeng Y., Li J., Wang C., Li W., He Z., et al. Phenotypical changes and clinical significance of CD4+/CD8+ T cells in SLE. Lupus Science & Medicine. 2022;9 doi: 10.1136/lupus-2022-000660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ma W.T., Gao F., Gu K., Chen D.K. The role of monocytes and macrophages in autoimmune diseases: a comprehensive review. Front. Immunol. 2019;10:1140. doi: 10.3389/fimmu.2019.01140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Vijayan D., Young A., Teng M., Smyth M.J. Targeting immunosuppressive adenosine in cancer. Nat. Rev. Cancer. 2017;17:709–724. doi: 10.1038/nrc.2017.86. [DOI] [PubMed] [Google Scholar]
- 7.Zhang Q., Li X., Yin X., Wang H., Fu C., Wang H., et al. Metabolomic profiling reveals serum L-pyroglutamic acid as a potential diagnostic biomarker for systemic lupus erythematosus. Rheumatology. 2021;60:598–606. doi: 10.1093/rheumatology/keaa126. [DOI] [PubMed] [Google Scholar]
- 8.Antonioli L., Colucci R., La Motta C., Tuccori M., Awwad O., Da Settimo F., et al. Adenosine deaminase in the modulation of immune system and its potential as a novel target for treatment of inflammatory disorders. Curr. Drug Targets. 2012;13:842–862. doi: 10.2174/138945012800564095. [DOI] [PubMed] [Google Scholar]
- 9.Sauer A.V., Morbach H., Brigida I., Ng Y.S., Aiuti A., Meffre E. Defective B cell tolerance in adenosine deaminase deficiency is corrected by gene therapy. J. Clin. Invest. 2012;122:2141–2152. doi: 10.1172/JCI61788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lee P.Y., Schulert G.S., Canna S.W., Huang Y., Sundel J., Li Y., et al. Adenosine deaminase 2 as a biomarker of macrophage activation syndrome in systemic juvenile idiopathic arthritis. Ann. Rheum. Dis. 2020;79:225–231. doi: 10.1136/annrheumdis-2019-216030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zavialov A.V., Engstrom A. Human ADA2 belongs to a new family of growth factors with adenosine deaminase activity. Biochem. J. 2005;391:51–57. doi: 10.1042/BJ20050683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kaljas Y., Liu C., Skaldin M., Wu C., Zhou Q., Lu Y., et al. Human adenosine deaminases ADA1 and ADA2 bind to different subsets of immune cells. Cell. Mol. Life Sci. 2017;74:555–570. doi: 10.1007/s00018-016-2357-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gao Z.W., Wang X., Zhang H.Z., Lin F., Liu C., Dong K. The roles of adenosine deaminase in autoimmune diseases. Autoimmun. Rev. 2021;20 doi: 10.1016/j.autrev.2020.102709. [DOI] [PubMed] [Google Scholar]
- 14.Ohta A. A metabolic immune checkpoint: adenosine in tumor microenvironment. Front. Immunol. 2016;7:109. doi: 10.3389/fimmu.2016.00109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Thien M., Phan T.G., Gardam S., Amesbury M., Basten A., Mackay F., et al. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity. 2004;20:785–798. doi: 10.1016/j.immuni.2004.05.010. [DOI] [PubMed] [Google Scholar]
- 16.Petri M., Stohl W., Chatham W., McCune W.J., Chevrier M., Ryel J., et al. Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008;58:2453–2459. doi: 10.1002/art.23678. [DOI] [PubMed] [Google Scholar]
- 17.Fernandez M.M., Grau G.E., Fernandez-Llanio C.N., Chalmeta V.I., Ivorra C.J., Castellano C.J., et al. Increased interferon-1alpha, interleukin-10 and BLyS concentrations as clinical activity biomarkers in systemic lupus erythematosus. Med. Clin. 2019;153:225–231. doi: 10.1016/j.medcli.2018.12.012. [DOI] [PubMed] [Google Scholar]
- 18.Huang S.P., Snedecor S.J., Nanji S., Lloyd E., Bell C.F. Real-world effectiveness of Belimumab in systemic lupus erythematosus: a systematic literature review. Rheumatol Ther. 2022 doi: 10.1007/s40744-022-00454-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Saze Z., Schuler P.J., Hong C.S., Cheng D., Jackson E.K., Whiteside T.L. Adenosine production by human B cells and B cell-mediated suppression of activated T cells. Blood. 2013;122:9–18. doi: 10.1182/blood-2013-02-482406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hesse J., Siekierka-Harreis M., Steckel B., Alter C., Schallehn M., Honke N., et al. Profound inhibition of CD73-dependent formation of anti-inflammatory adenosine in B cells of SLE patients. EBioMedicine. 2021;73 doi: 10.1016/j.ebiom.2021.103616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Knight J.S., Mazza L.F., Yalavarthi S., Sule G., Ali R.A., Hodgin J.B., et al. Ectonucleotidase-mediated suppression of lupus autoimmunity and vascular dysfunction. Front. Immunol. 2018;9:1322. doi: 10.3389/fimmu.2018.01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dang J., Xu Z., Xu A., Liu Y., Fu Q., Wang J., et al. Human gingiva-derived mesenchymal stem cells are therapeutic in lupus nephritis through targeting of CD39(-)CD73 signaling pathway. J. Autoimmun. 2020;113 doi: 10.1016/j.jaut.2020.102491. [DOI] [PubMed] [Google Scholar]
- 23.Levack R.C., Newell K.L., Cabrera-Martinez B., Cox J., Perl A., Bastacky S.I., Winslow G.M. Adenosine receptor 2a agonists target mouse CD11c+T-bet+ B cells in infection and autoimmunity. Nat. Commun. 2022;13:452. doi: 10.1038/s41467-022-28086-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bortoluzzi A., Vincenzi F., Govoni M., Padovan M., Ravani A., Borea P.A., Varani K. A2A adenosine receptor upregulation correlates with disease activity in patients with systemic lupus erythematosus. Arthritis Res. Ther. 2016;18:192. doi: 10.1186/s13075-016-1089-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tretter T., Venigalla R.K., Eckstein V., Saffrich R., Sertel S., Ho A.D., et al. Induction of CD4+ T-cell anergy and apoptosis by activated human B cells. Blood. 2008;112:4555–4564. doi: 10.1182/blood-2008-02-140087. [DOI] [PubMed] [Google Scholar]
- 26.Yan B., Ye S., Chen G., Kuang M., Shen N., Chen S. Dysfunctional CD4+,CD25+ regulatory T cells in untreated active systemic lupus erythematosus secondary to interferon-alpha-producing antigen-presenting cells. Arthritis Rheum. 2008;58:801–812. doi: 10.1002/art.23268. [DOI] [PubMed] [Google Scholar]
- 27.Atfy M., Amr G.E., Elnaggar A.M., Labib H.A., Esh A., Elokely A.M. Impact of CD4+CD25high regulatory T-cells and FoxP3 expression in the peripheral blood of patients with systemic lupus erythematosus. Egypt. J. Immunol. 2009;16:117–126. [PubMed] [Google Scholar]
- 28.Suen J.L., Li H.T., Jong Y.J., Chiang B.L., Yen J.H. Altered homeostasis of CD4(+) FoxP3(+) regulatory T-cell subpopulations in systemic lupus erythematosus. Immunology. 2009;127:196–205. doi: 10.1111/j.1365-2567.2008.02937.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tselios K., Sarantopoulos A., Gkougkourelas I., Boura P. CD4+CD25highFOXP3+ T regulatory cells as a biomarker of disease activity in systemic lupus erythematosus: a prospective study. Clin. Exp. Rheumatol. 2014;32:630–639. [PubMed] [Google Scholar]
- 30.Zabinska M., Krajewska M., Koscielska-Kasprzak K., Jakuszko K., Bartoszek D., Myszka M., et al. CD4(+)CD25(+)CD127(-) and CD4(+)CD25(+)Foxp3(+) regulatory T cell subsets in mediating autoimmune reactivity in systemic lupus erythematosus patients. Arch. Immunol. Ther. Exp. 2016;64:399–407. doi: 10.1007/s00005-016-0399-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Handono K., Firdausi S.N., Pratama M.Z., Endharti A.T., Kalim H. Vitamin A improve Th17 and Treg regulation in systemic lupus erythematosus. Clin. Rheumatol. 2016;35:631–638. doi: 10.1007/s10067-016-3197-x. [DOI] [PubMed] [Google Scholar]
- 32.Lin S.C., Chen K.H., Lin C.H., Kuo C.C., Ling Q.D., Chan C.H. The quantitative analysis of peripheral blood FOXP3-expressing T cells in systemic lupus erythematosus and rheumatoid arthritis patients. Eur. J. Clin. Invest. 2007;37:987–996. doi: 10.1111/j.1365-2362.2007.01882.x. [DOI] [PubMed] [Google Scholar]
- 33.Lyssuk E.Y., Torgashina A.V., Soloviev S.K., Nassonov E.L., Bykovskaia S.N. Reduced number and function of CD4+CD25highFoxP3+ regulatory T cells in patients with systemic lupus erythematosus. Adv. Exp. Med. Biol. 2007;601:113–119. [PubMed] [Google Scholar]
- 34.Zhao S.S., Li X.M., Li X.P., Zhai Z.M., Chen Z., Ma Y., et al. [Expression of CD4+ CD25+ CD127(low/-) T cells in patients with systemic lupus erythematosus. Zhonghua Yixue Zazhi. 2008;88:453–456. [PubMed] [Google Scholar]
- 35.Pan X., Yuan X., Zheng Y., Wang W., Shan J., Lin F., et al. Increased CD45RA+ FoxP3(low) regulatory T cells with impaired suppressive function in patients with systemic lupus erythematosus. PLoS One. 2012;7 doi: 10.1371/journal.pone.0034662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Szmyrka-Kaczmarek M., Kosmaczewska A., Ciszak L., Szteblich A., Wiland P. Peripheral blood Th17/Treg imbalance in patients with low-active systemic lupus erythematosus. Postepy Hig. Med. Dosw. 2014;68:893–898. doi: 10.5604/17322693.1111127. [DOI] [PubMed] [Google Scholar]
- 37.El-Maraghy N., Ghaly M.S., Dessouki O., Nasef S.I., Metwally L. CD4+CD25-Foxp3+ T cells as a marker of disease activity and organ damage in systemic lupus erythematosus patients. Arch. Med. Sci. 2018;14:1033–1040. doi: 10.5114/aoms.2016.63597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kamal M., Gabr H., Anwar S., Bastawy S., Salah L. The relation of CD4(+)CD25+Foxp3+ regulatory T cells concentration with disease activity and damage index in systemic lupus erythematosus. Lupus. 2022;31:463–471. doi: 10.1177/09612033221083269. [DOI] [PubMed] [Google Scholar]
- 39.Valencia X., Yarboro C., Illei G., Lipsky P.E. Deficient CD4+CD25high T regulatory cell function in patients with active systemic lupus erythematosus. J. Immunol. 2007;178:2579–2588. doi: 10.4049/jimmunol.178.4.2579. [DOI] [PubMed] [Google Scholar]
- 40.Yuliasih Y., Rahmawati L.D., Putri R.M. Th17/Treg ratio and disease activity in systemic lupus erythematosus. Caspian J Intern Med. 2019;10:65–72. doi: 10.22088/cjim.10.1.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhao C., Chu Y., Liang Z., Zhang B., Wang X., Jing X., et al. Low dose of IL-2 combined with rapamycin restores and maintains the long-term balance of Th17/Treg cells in refractory SLE patients. BMC Immunol. 2019;20:32. doi: 10.1186/s12865-019-0305-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chu Y., Zhao C., Zhang B., Wang X., Wang Y., An J., et al. Restoring T-helper 17 cell/regulatory T-cell balance and decreasing disease activity by rapamycin and all-trans retinoic acid in patients with systemic lupus erythematosus. Lupus. 2019;28:1397–1406. doi: 10.1177/0961203319877239. [DOI] [PubMed] [Google Scholar]
- 43.Fu D., Senouthai S., Wang J., You Y. Vasoactive intestinal peptide ameliorates renal injury in a pristane-induced lupus mouse model by modulating Th17/Treg balance. BMC Nephrol. 2019;20:350. doi: 10.1186/s12882-019-1548-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Smyth L.A., Ratnasothy K., Tsang J.Y., Boardman D., Warley A., Lechler R., et al. CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur. J. Immunol. 2013;43:2430–2440. doi: 10.1002/eji.201242909. [DOI] [PubMed] [Google Scholar]
- 45.Jarvis L.B., Rainbow D.B., Coppard V., Howlett S.K., Georgieva Z., Davies J.L., et al. Therapeutically expanded human regulatory T-cells are super-suppressive due to HIF1A induced expression of CD73. Commun. Biol. 2021;4:1186. doi: 10.1038/s42003-021-02721-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bono M.R., Fernández D., Flores-Santibáñez F., Rosemblatt M., Sauma D. CD73 and CD39 ectonucleotidases in T cell differentiation: beyond immunosuppression. FEBS Lett. 2015;589:3454–3460. doi: 10.1016/j.febslet.2015.07.027. [DOI] [PubMed] [Google Scholar]
- 47.Shore A., Dosch H.M., Gelfand E.W. Role of adenosine deaminase in the early stages of precursor T cell maturation. Clin. Exp. Immunol. 1981;44:152–155. [PMC free article] [PubMed] [Google Scholar]
- 48.Naval-Macabuhay I., Casanova V., Navarro G., Garcia F., Leon A., Miralles L., et al. Adenosine deaminase regulates Treg expression in autologous T cell-dendritic cell cocultures from patients infected with HIV-1. J. Leukoc. Biol. 2016;99:349–359. doi: 10.1189/jlb.3A1214-580RR. [DOI] [PubMed] [Google Scholar]
- 49.Tardif V., Muir R., Cubas R., Chakhtoura M., Wilkinson P., Metcalf T., et al. Adenosine deaminase-1 delineates human follicular helper T cell function and is altered with HIV. Nat. Commun. 2019;10:823. doi: 10.1038/s41467-019-08801-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Naval-Macabuhay I., Casanova V., Navarro G., Garcia F., Leon A., Miralles L., et al. Adenosine deaminase regulates Treg expression in autologous T cell-dendritic cell cocultures from patients infected with HIV-1. J. Leukoc. Biol. 2016;99:349–359. doi: 10.1189/jlb.3A1214-580RR. [DOI] [PubMed] [Google Scholar]
- 51.Tardif V., Muir R., Cubas R., Chakhtoura M., Wilkinson P., Metcalf T., et al. Adenosine deaminase-1 delineates human follicular helper T cell function and is altered with HIV. Nat. Commun. 2019;10:823. doi: 10.1038/s41467-019-08801-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wu Z., Gao S., Watanabe N., Batchu S., Kajigaya S., Diamond C., et al. Single-cell profiling of T lymphocytes in deficiency of adenosine deaminase 2. J. Leukoc. Biol. 2022;111:301–312. doi: 10.1002/JLB.5A0621-314R. [DOI] [PubMed] [Google Scholar]
- 53.Ohta A., Kini R., Ohta A., Subramanian M., Madasu M., Sitkovsky M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 2012;3:190. doi: 10.3389/fimmu.2012.00190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ohta A., Sitkovsky M. Extracellular adenosine-mediated modulation of regulatory T cells. Front. Immunol. 2014;5:304. doi: 10.3389/fimmu.2014.00304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ehrentraut H., Westrich J.A., Eltzschig H.K., Clambey E.T. Adora2b adenosine receptor engagement enhances regulatory T cell abundance during endotoxin-induced pulmonary inflammation. PLoS One. 2012;7 doi: 10.1371/journal.pone.0032416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Baumann I., Kolowos W., Voll R.E., Manger B., Gaipl U., Neuhuber W.L., et al. Impaired uptake of apoptotic cells into tingible body macrophages in germinal centers of patients with systemic lupus erythematosus. Arthritis Rheum. 2002 Jan;46(1):191–201. doi: 10.1002/1529-0131(200201)46:1<191::AID-ART10027>3.0.CO;2-K. 10.1002/1529-0131(200201)46:1<191::AID-ART10027>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
- 57.Tas S.W., Quartier P., Botto M., Fossati-Jimack L. Macrophages from patients with SLE and rheumatoid arthritis have defective adhesion in vitro, while only SLE macrophages have impaired uptake of apoptotic cells. Ann. Rheum. Dis. 2006;65:216–221. doi: 10.1136/ard.2005.037143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bijl M., Reefman E., Horst G., Limburg P.C., Kallenberg C.G. Reduced uptake of apoptotic cells by macrophages in systemic lupus erythematosus: correlates with decreased serum levels of complement. Ann. Rheum. Dis. 2006;65:57–63. doi: 10.1136/ard.2005.035733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Labonte A.C., Kegerreis B., Geraci N.S., Bachali P., Madamanchi S., Robl R., et al. Identification of alterations in macrophage activation associated with disease activity in systemic lupus erythematosus. PLoS One. 2018;13 doi: 10.1371/journal.pone.0208132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Horuluoglu B., Bayik D., Kayraklioglu N., Goguet E., Kaplan M.J., Klinman D.M. PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J. Autoimmun. 2019;99:24–32. doi: 10.1016/j.jaut.2019.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Mohammadi S., Saghaeian-Jazi M., Sedighi S., Memarian A. Immunomodulation in systemic lupus erythematosus: induction of M2 population in monocyte-derived macrophages by pioglitazone. Lupus. 2017;26:1318–1327. doi: 10.1177/0961203317701842. [DOI] [PubMed] [Google Scholar]
- 62.Lourenco E.V., Wong M., Hahn B.H., Palma-Diaz M.F., Skaggs B.J. Laquinimod delays and suppresses nephritis in lupus-prone mice and affects both myeloid and lymphoid immune cells. Arthritis Rheumatol. 2014;66:674–685. doi: 10.1002/art.38259. [DOI] [PubMed] [Google Scholar]
- 63.Li F., Yang Y., Zhu X., Huang L., Xu J. Macrophage polarization modulates development of systemic lupus erythematosus. Cell. Physiol. Biochem. 2015;37:1279–1288. doi: 10.1159/000430251. [DOI] [PubMed] [Google Scholar]
- 64.Horuluoglu B., Bayik D., Kayraklioglu N., Goguet E., Kaplan M.J., Klinman D.M. PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J. Autoimmun. 2019;99:24–32. doi: 10.1016/j.jaut.2019.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Csoka B., Selmeczy Z., Koscso B., Nemeth Z.H., Pacher P., Murray P.J., et al. Adenosine promotes alternative macrophage activation via A2A and A2B receptors. Faseb. J. 2012;26:376–386. doi: 10.1096/fj.11-190934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zavialov A.V., Gracia E., Glaichenhaus N., Franco R., Zavialov A.V., Lauvau G. Human adenosine deaminase 2 induces differentiation of monocytes into macrophages and stimulates proliferation of T helper cells and macrophages. J. Leukoc. Biol. 2010;88:279–290. doi: 10.1189/jlb.1109764. [DOI] [PubMed] [Google Scholar]
- 67.Tiwari-Heckler S., Yee E.U., Yalcin Y., Park J., Nguyen D.T., Gao W., et al. Adenosine deaminase 2 produced by infiltrative monocytes promotes liver fibrosis in nonalcoholic fatty liver disease. Cell Rep. 2021;37 doi: 10.1016/j.celrep.2021.109897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhu C., Mustafa D., Zheng P.P., van der Weiden M., Sacchetti A., Brandt M., et al. Activation of CECR1 in M2-like TAMs promotes paracrine stimulation-mediated glial tumor progression. Neuro Oncol. 2017;19:648–659. doi: 10.1093/neuonc/now251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Xu S., Zhu W., Shao M., Zhang F., Guo J., Xu H., et al. Ecto-5'-nucleotidase (CD73) attenuates inflammation after spinal cord injury by promoting macrophages/microglia M2 polarization in mice. J. Neuroinflammation. 2018;15:155. doi: 10.1186/s12974-018-1183-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Costales M.G., Alam M.S., Cavanaugh C., Williams K.M. Extracellular adenosine produced by ecto-5'-nucleotidase (CD73) regulates macrophage pro-inflammatory responses, nitric oxide production, and favors Salmonella persistence. Nitric Oxide. 2018;72:7–15. doi: 10.1016/j.niox.2017.11.001. [DOI] [PubMed] [Google Scholar]
- 71.Gill M.A., Blanco P., Arce E., Pascual V., Banchereau J., Palucka A.K. Blood dendritic cells and DC-poietins in systemic lupus erythematosus. Hum. Immunol. 2002;63:1172–1180. doi: 10.1016/s0198-8859(02)00756-5. [DOI] [PubMed] [Google Scholar]
- 72.Chan V.S., Nie Y.J., Shen N., Yan S., Mok M.Y., Lau C.S. Distinct roles of myeloid and plasmacytoid dendritic cells in systemic lupus erythematosus. Autoimmun. Rev. 2012;11:890–897. doi: 10.1016/j.autrev.2012.03.004. [DOI] [PubMed] [Google Scholar]
- 73.Blanco P., Palucka A.K., Gill M., Pascual V., Banchereau J. Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science. 2001;294:1540–1543. doi: 10.1126/science.1064890. [DOI] [PubMed] [Google Scholar]
- 74.Palucka A.K., Banchereau J., Blanco P., Pascual V. The interplay of dendritic cell subsets in systemic lupus erythematosus. Immunol. Cell Biol. 2002;80:484–488. doi: 10.1046/j.1440-1711.2002.01112.x. [DOI] [PubMed] [Google Scholar]
- 75.Silva-Vilches C., Ring S., Mahnke K. ATP and its metabolite adenosine as regulators of dendritic cell activity. Front. Immunol. 2018;9:2581. doi: 10.3389/fimmu.2018.02581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Novitskiy S.V., Ryzhov S., Zaynagetdinov R., Goldstein A.E., Huang Y., Tikhomirov O.Y., et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood. 2008;112:1822–1831. doi: 10.1182/blood-2008-02-136325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liu C., Shang Q., Bai Y., Guo C., Zhu F., Zhang L., et al. Adenosine A2A receptor, a potential valuable target for controlling reoxygenated DCs-triggered inflammation. Mol. Immunol. 2015;63:559–565. doi: 10.1016/j.molimm.2014.10.012. [DOI] [PubMed] [Google Scholar]
- 78.Silva-Vilches C., Ring S., Schrader J., Clausen B.E., Probst H.C., Melchior F., et al. Production of extracellular adenosine by CD73(+) dendritic cells is crucial for induction of tolerance in contact hypersensitivity reactions. J. Invest. Dermatol. 2019;139:541–551. doi: 10.1016/j.jid.2018.10.016. [DOI] [PubMed] [Google Scholar]
- 79.Gary E., O'Connor M., Chakhtoura M., Tardif V., Kumova O.K., Malherbe D.C., et al. Adenosine deaminase-1 enhances germinal center formation and functional antibody responses to HIV-1 Envelope DNA and protein vaccines. Vaccine. 2020;38:3821–3831. doi: 10.1016/j.vaccine.2020.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
There is no research related data stored in publicly available repositories, and the data in this paper will be made available on request from the corresponding author.



