Skip to main content

REVIEW article

Front. Immunol., 06 January 2025
Sec. Autoimmune and Autoinflammatory Disorders : Autoimmune Disorders

An updated review on abnormal epigenetic modifications in the pathogenesis of systemic lupus erythematosus

  • Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. The inconsistent prevalence of SLE between monozygotic twins suggests that environmental factors affect the occurrence of this disease. Abnormal epigenetic regulation is strongly associated with the pathogenesis of SLE. Epigenetic mechanisms may be involved in the development of lupus through DNA methylation, histone modification, noncoding RNAs, and other modifications. This review aims to show numerous studies as a treasure map to better understand the effects of aberrant epigenetic modification in the onset and development of SLE, which will benefit the current basic research and provide potential diagnostic biomarkers or therapeutic targets for SLE.

1 Introduction

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of multiple autoantibodies. The produced autoantibodies bind to autoantigens as immune complexes, circulating in the body, depositing in various tissues, and causing chronic inflammation (1). Failure of self-tolerance is considered the main pathogenesis of SLE, causing dysregulation in both the innate and adaptive immune systems. The upstream regulatory mechanisms that determine immune dysfunctions have been extensively documented. Based on genome-wide association studies, many scholars have attributed the pathogenesis of lupus to genetic susceptibility. Through the study of familial SLE, researchers have identified multiple loci of SLE susceptibility (2). However, the consistency of monozygotic twins with lupus is only 24–57% (3). Meanwhile, epidemiological studies of environmental exposure have shown that drugs and ultraviolet light can trigger lupus-like disease (4), suggesting that both environmental factors and genetic predisposition contribute to the development of SLE. For decades, an increasing number of studies have revealed that environmental factors play regulatory roles through epigenetic mechanisms contributing to the development of SLE, indicating that epigenetic regulation is an important contributing factor in SLE (5). This review aims to present recent advances in epigenetic factors involved in the pathogenesis, biomarkers, and therapeutic targets of SLE to facilitate an understanding of the effects of epigenetic abnormalities in SLE pathogenesis.

2 Epigenetic alterations in SLE

Epigenetic changes, including DNA methylation, histone modifications, noncoding RNAs (ncRNAs), and RNA methylation, are thought to be key signaling mediators between the genome and the environment. Richardson et al. reported that CD4+ T cells were observed to increase self-reactivity when they were treated with a DNA methylation inhibitor, 5-azacytidine (6). Since then, a series of studies have identified the function of DNA demethylation in SLE.

2.1 DNA methylation in SLE

DNA methylation is a dynamic process that involves both methylation and demethylation events (7). Methylation acts as a transcriptional repressive modification, which is defined as the addition of a methyl group to the C5 position of cytosine in CpG dinucleotides by DNA methyltransferase (DNMTs). The methyl groups interfere with the binding of transcription factors to DNA, thereby partially causing the silencing of those genes. Abnormal methylation patterns lead to aberrant gene activation, which contributes to SLE development. 5-Hydroxymethylcytosine (5-hmC) regulates gene transcription, resulting in dysregulation of the immune system in SLE (8). The modification of 5-hmC was found for the first time in the DNA of bacteriophages (9). Hydroxymethylation of 5-methylcytosine (5-mC) produces 5-hmC, which is further oxidized to 5-formylcytosine (5-fC) and 5-carboxycytosine (5-caC) (10). The stepwise oxidation of 5-mC is a demethylation mechanism which activates gene transcription, promotes gene expression and is catalyzed by ten-eleven translocation (TET), a methylcytosine dioxygenase (11) (Figure 1).

Figure 1
www.frontiersin.org

Figure 1. Epigenetic mechanisms in SLE. (A) DNA methylation: DNMTs add methyl groups to the C5 position of cytosine in CpG dinucleotides, forming 5mC. This modification silences genes by interfering with transcription factor binding. TET enzymes reverse this by converting 5mC to 5hmC, the first step in the DNA demethylation process. (B) Histone modifications: Modifications such as acetylation, methylation, phosphorylation, and ubiquitination target specific amino acids in histone tails, altering chromatin structure and gene expression. Histone acetyltransferases (HATs) add acetyl groups to lysine residues, activating transcription, while HDACs remove acetyl groups, repressing transcription. Histone methyltransferases (HMTs) and demethylases (HDMs) respectively add or remove methyl groups on lysine and arginine residues. (C) Noncoding RNAs: MiRNAs regulate gene expression by binding to complementary sequences in the 3′ untranslated region (UTR) of target mRNAs, promoting degradation or inhibiting translation. LncRNAs regulate transcription by interacting with proteins such as transcription factors, and influence translation by binding to mRNA. Both lncRNAs and circRNAs act as miRNA sponges, influencing transcriptional and post-transcriptional gene regulation. (D) RNA methylation: RNA methylation is regulated by three groups of enzymes: “writers” (methyltransferases), “erasers” (demethylases), and “readers” (m6A-binding proteins). This process modifies RNA function post-transcriptionally. Ac, acetyl group; eIF3, eukaryotic initiation factor 3; Me, methyl group; m6RNA, N6-methyladenosine.

Many signaling pathways, transcription factors, and ncRNAs have been proven to affect DNA methylation patterns in CD4+ T cells, B cells, monocytes, neutrophils, and dendritic cells as well. DNA hypomethylation plays an important role in the pathogenesis of SLE. Studies have revealed that the hypomethylation of global genomic DNA and many immune-related genes in SLE CD4+ T cells result in the overexpression of growth arrest and DNA damage inducible 45 alpha (Gadd45a), CD70, CD11a, CD40L, and perforin, thereby contributing to autoimmunity (1217). Zhao et al. found that 5-hmC was increased in SLE CD4+ T cells, which indicated that DNA hydroxymethylation was involved in the aberrant regulation of gene transcription in SLE pathogenesis (8). The following are the applications of DNA methylation in SLE reported over the past 2 years. More classical pathogenic mechanisms of DNA methylation in SLE over the past 5 years are listed in Table 1.

Table 1
www.frontiersin.org

Table 1. Altered DNA methylation genes in SLE (over the past 5 years).

Liao et al. found that the decreased mRNA expression and serum concentration of TNF-like weak inducer of apoptosis (TWEAK) significantly correlated with SLE Disease Activity Index (SLEDAI) and renal damage in SLE patients. Increased DNA methylation levels of TWEAK in the peripheral blood of SLE patients suggested that abnormal DNA methylation may participate in SLE pathogenesis by downregulating the expression of TWEAK (18). Local DNA methylation of the CD45 gene was considered to participate in the regulation of CD45 isoform expression in SLE peripheral blood mononuclear cells (PBMCs) (19). Gao reported that in identifying feature autophagy-related genes (ARGs) in SLE PBMCs, the cg24898863 (S100A8) gene was hypomethylated and upregulated, whereas the cg27490128 (NCR3) gene was hypermethylated and downregulated, prompting the possible mechanism of ARGs involved in the process of SLE (20).

Luo et al. found that the signal transducer and activator of transcription 3 (STAT3) interacted with TET2, which induced DNA demethylation of the interferon-inducible 44 like (IFI44L) promoter. Overexpression of IFI44L in monocytes can upregulate co-stimulatory receptors, induce Th1/Th17-related cytokines, and promote maturation of monocyte-derived dendritic cells (21), thus leading to autoimmunity in SLE. Hurtado et al. also reported hypomethylation of the IFI44L gene in resting naive B cells of SLE patients, suggesting that epigenetic alterations are established very early in B-cell ontogeny (22). It has been shown that the conserved noncoding sequence 2 region of forkhead box protein 3 (FoxP3) was hypermethylation in SLE follicular regulatory T cells, leading to transcriptional suppression and functional decline of FoxP3 (23). Liu et al. found that downregulation of ubiquitin-like with PHD and RING finger domains 1 (UHRF1) in the T follicular helper (Tfh) cells of SLE patients, decreased UHRF1 can reduce DNA methylation and H3K27me3 levels in the B cell lymphoma 6 (BCL6) promoter region, which resulted in the increased level of BCL6 and accelerated differentiation of Tfh cells (24). The result revealed the role of UHRF1 in regulating Tfh cell differentiation and provided a potential therapeutic target for SLE.

Recent studies have shown that the matrix metallopeptidase 9 (MMP9) promoter methylation level is significantly reduced while the runt-related transcription factor 3 (RUNX3) promoter methylation level is significantly heightened in SLE PBMCs compared to healthy controls. According to receiver operating characteristic (ROC) analysis, the diagnostic efficiency of the MMP9 promoter methylation level for SLE was 0.839 while the RUNX3 promoter methylation level for SLE was 0.769, emphasizing the potential utility of MMP9 and RUNX3 methylation levels as biomarkers for SLE diagnosis (25, 26). In the lupus nephritis (LN) group compared to both the SLE group without kidney injury and healthy controls, cg08332381 and cg03297029 were significantly hypermethylated while cg16797344 was significantly hypomethylated. According to ROC curve analysis, the diagnostic efficiency of these sites for LN was approximately 0.8, and the combined efficiency of all three sites exceeded 0.9, which emphasizes the potential use of cg08332381, cg03297029, and cg16797344 methylation levels as biomarkers for LN diagnosis (27).

Methyl-CpG binding protein 2 (MeCP2) selectively binds to 5-mC residues in CpG dinucleotides to regulate gene expression. Li et al. recently demonstrated that the overexpression of MeCP2 is markedly linked to an elevation in brain-derived neurotrophic factor (BDNF), potentially leading to disturbances in normal neuronal function in mice. C57BL/6 transgenic mice with human MeCP2 (B6.Mecp2Tg1) exhibit lupus-like phenotypes and significant central nervous system (CNS) dysfunction, making them a potential model for neuropsychiatric lupus (NPSLE) (28).

2.2 Histone modifications in SLE

Chromatin is composed of nucleosomes, histone octamers, and the surrounding DNA. Various modifications, such as acetylation, phosphorylation, ubiquitination, and methylation, target specific amino acids in histone tails (33), affecting the structure of chromatin and thus regulating post-translational gene expression. H3 lysine 9 trimethylation (H3K9me3) results in transcriptional inhibition (34), whereas H3 lysine 4 trimethylation (H3K4me3) is associated with transcriptional activation (35) (Figure 1).

In comparison with healthy controls, B cells have been observed in global histone 3 (H3) and histone 4 (H4) hypoacetylation in SLE patients (36). Zhang et al. found that in the miR-1246 promoter region of SLE B cells, H3 lysine 27 trimethylation (H3K27me3) was increased, histone H3 acetylation at Lys9 and Lys14 (H3K9/K14ac) was reduced, and downregulated miR-1246 expression led to B-cell hyperactivity (37).

Luo et al. reported that in CD4+ T cells of SLE, reduced suppressor of variation 3–9 homolog 1 (SUV39H1) in the cAMP-responsive element modulator α (CREMα) promoter region resulted in decreased H3K9me3 levels. The SET domain containing 1 (Set1) expression in the CREMα promoter region was increased. These resulted in decreased DNMT3a and DNA methylation levels and increased H3K4me3 levels, which promoted CREMα transcription and ultimately SLE (38). Studies have found that the reduction of jumonji domain-containing 3 (JMJD3) at the hematopoietic progenitor kinase 1 (HPK1) promoter increased H3K27me3, leading to the reduced mixed-lineage leukemia 1 and H3K4me3 abundance in Tfh cells of SLE patients. All of these resulted in HPK1 low expression and Tfh cell overactivation, ultimately inducing the development of SLE (39). Liu et al. reported that elevated M2-like phenotype in SLE monocytes was regulated by elevated acetylation levels of H3 in proliferator-activated receptor-γ (PPAR-γ) promoter. Due to the immunosuppressive function of M2-like monocytes, this study may propose a potential treatment for SLE patients (40).

There is limited research on the impact of histone modifications on the pathogenesis of NPSLE. Recent studies have found that the overexpression of MeCP2 is significantly associated with the upregulation of nuclear receptor corepressor 1 (NCoR1) and histone deacetylases (HDACs). This transcriptional dysregulation may further contribute to the development of lupus-like phenotypes in mice, along with significant central nervous system (CNS) dysfunction (28).

In this study, we present aberrant histone modifications in immune cells from SLE patients during the past 2 years. More classic studies over the past 5 years are listed in Table 2.

Table 2
www.frontiersin.org

Table 2. Altered histone modifications in SLE (over the past 5 years).

2.3 Noncoding RNAs in SLE

The human genome is extensively transcribed, and over 80% of RNA transcripts are ncRNAs, which do not translate into proteins (44). NcRNAs are the most recently discovered epigenetic mechanisms (Figure 1). MicroRNAs (miRNAs), short noncoding RNAs, are single-stranded RNAs of 18–22 nucleotides emerging as key posttranscriptional regulators of target genes by degrading mRNAs and repressing their translation (45). Long noncoding RNAs (lncRNAs) have a length of over 200 nucleotides (46). Circular RNA (circRNA) is a type of single-stranded RNA formed as a covalently closed continuous loop. Identifying useful biomarkers for early diagnosis and treatment of SLE is a challenge in current research.

2.3.1 MicroRNAs in SLE

MiRNA binds to target genes at the complementary loci in the 3′ untranslated region to regulate expression by degrading mRNA and inhibiting translation (47). MiRNA regulation plays a pivotal role in many biological processes and diseases, such as cancer and autoimmune diseases, including SLE (48).

Luo et al. found that the expression of miR-301a-3p is significantly increased in SLE PBMCs, promoting the expression of IL-6, IL-17, and interferon-γ (INF-γ), as well as IL-1 receptor-associated kinase 1 (IRAK1)-mediated Th17 cell differentiation by targeting Pellino 1 (Peli1) (49). Downregulated expression of miR-99a-3p can induce B-cell autophagy through its target gene eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1)-mediated autophagy signaling pathway in SLE B cells (50). In SLE CD4+ T cells, downregulated miR-124 promotes immunoactivity by upregulating interferon regulatory factor 1 (51). In addition, upregulated miR-152-3p in SLE CD4+ T cells was involved in the development of SLE by targeting DNMT1 to inhibit myeloid differentiation factor 88 (MyD88) methylation and promote toll-like receptor (TLR)-mediated cellular inflammatory responses (52). In terms of innate immunity, upregulation of miR-210-5p in macrophages from SLE patients can inhibit specificity protein 1 (SP1)- and HSCARG-mediated NADPH oxidase (NOX) activity and reactive oxygen species production, leading to the accumulation of secondary necrotic cells, which is involved in the pathogenesis of SLE (53).

Chen et al. constructed a multi-miRNA detection platform based on target-triggered locked hairpin DNA-functionalized Au nanoprobes for the diagnosis and classification of SLE. The results showed that the area under curve (AUC) value for the combined signature of three urinary small extracellular vesicle (sEV) miRNAs (miR-146a, miR-29c, and miR-150) reached 1.00 (54). ElFeky et al. reported that the expression of miR-199a, miR-21, and miR-146a was significantly increased in the serum of LN patients compared to healthy controls (HCs) and SLE patients without LN. According to ROC curve analysis, the AUC values of miR-199a, miR-21, and miR-146a in distinguishing LN from SLE patients without LN were 0.96, 0.82 and 0.90, respectively. Logistic regression analysis showed that miR-199a was an independent predictor of LN with an OR of 1.69 (55).

A recent study reported that miR-155 was upregulated in regulatory T cells (Tregs) from SLE and SLE-induced mice. Inflammation-induced miR-155 impaired Treg function by decreasing suppressor of cytokine signaling-1 (SOCS1) expression. In the SLE-induced mice model, miR-155 inhibition improved Treg function under inflammatory stimulation and alleviated SLE (56). Wu et al. found that the expression of miR-125b-5p was upregulated in SLE PBMCs. After treatment with umbilical cord mesenchymal stem cells (UC-MSCs) modified by miR-125b-5p in MRL/lpr mice, the serum levels of IL-4 were elevated while IL-17A levels were reduced, and inflammatory infiltration and microthrombus formation in the lungs and kidneys were decreased (57). Targeting miR-155 and miR-125b-5p offers therapeutic approaches for alleviating SLE.

The above shows the role of miRNAs in SLE during the past 2 years. More classic studies over the past 5 years are listed in Table 3.

Table 3
www.frontiersin.org

Table 3. MicroRNA alteration in SLE (over the past 5 years).

2.3.2 Long noncoding RNA in SLE

LncRNAs participate in a variety of biological processes, such as silencing transcription, associating with proteins, activating protein-coding genes, binding to mRNAs, and acting as competing endogenous RNAs (ceRNAs) (83). LncRNAs have been identified to have a pivotal part in several pathogenic disorders (84) and are involved in autoimmune responses.

Nuclear enriched abundant transcript 1 (NEAT1), the best-characterized lncRNA, is known to play a crucial part in the innate immune response (85, 86). It was found that the upregulation of NEAT1 in monocyte-derived dendritic cells of SLE patients induced the expression of IL-6 and positively correlated with SLEDAI (87). Jiang et al. reported that the increased expression of NEAT1 in SLE PBMCs was negatively correlated with Th1/Th2 balance, which participated in the pathogenesis of SLE (88). LncNEAT1 contained two transcripts, lncNEAT1_1 and lncNEAT1_2, both of which were upregulated in the peripheral blood of childhood-onset SLE (cSLE). The lncNEAT1_2 expression was positively correlated with SLEDAI, fever, renal involvement, elevated ESR, and low C3 levels. According to ROC curve analysis, the AUC value was 0.812, with 62.2% sensitivity and 92.5% specificity (89).

Xiao et al. found that the expression of lncRNA growth arrest-specific transcript 5 (GAS5) was decreased in monocytes of SLE patients and negatively correlated with SLEDAI. GAS5 may be involved in TLR4-mediated inflammatory processes by inhibiting the activation of the mitogen activated protein kinase (MAPK) pathway, thus participating in the pathogenesis of SLE (90). It was also reported that lncRNA GAS5 expression was downregulated in SLE PBMCs and may contribute to SLE by targeting phosphatase and tensin homolog (PTEN) through competitively binding to miR-21 (91).

IL21 anti-sense RNA 1 (IL21-AS1) is a lncRNA located on the antisense strand of the IL21 gene motif. Liu et al. found that IL21-AS1 expression was upregulated in CD4+ T cells and Tfh cells from SLE patients and positively correlated with SLEDAI. Moreover, the increased acetylation levels of histone H3 on the IL21 promoter led to transcriptional activation of IL21 (92). However, a previous study reported that IL21-AS1 expression was downregulated in SLE CD4+ T cells and negatively correlated with SLEDAI, which may influence disease activity by participating in IL-2-mediated follicular regulatory T-cell activation in SLE (93). The different genetic backgrounds and the limited sample size in the previous study may explain the contrasting results of the two studies.

In both the serum and PBMCs of SLE patients, the expression of lncRNA H19 was increased, while that of miR-19b was decreased. Furthermore, according to ROC curve analysis, the AUC value of serum H19 was 0.853 for SLE diagnosis. Upregulation of H19 promoted apoptosis and the inflammatory response of PBMCs by interacting with miR-19b, which may participate in the pathogenesis of SLE (94). A recent study constructed a ceRNA network combined with clinical validation to screen for potential molecular markers of SLE through bioinformatics analysis. The results found that lncRNA X inactive specific transcript (XIST) and signal transducer and activator of transcription 1 (STAT1) were upregulated, while miR-381-3p was downregulated, in the peripheral blood of SLE. ROC curve analysis suggested that the lncRNA XIST/miR-381-3P/STAT1 axis could serve as a molecular marker for SLE diagnosis and a potential therapeutic target for LN (76). Liu et al. reported that lncRNA highly accelerated region 1 A (HAR1A) was significantly upregulated in PBMCs from LN patients and that it bound to miR-149-3p to upregulate SWItch/sucrose non-fermentable-related matrix-associated actin-dependent regulator of chromatin subfamily D member 1 (SMARCD1). The HAR1A/miR-149-3p/SMARCD1 pathway upregulated the expression of inducible nitric oxide synthase (iNOS), an inflammation inducer. Additionally, their study found that IL-10 secreted by iTreg cells alleviated LN through downregulating lncRNA HAR1A transcription, thereby suppressing SMARCD1-mediated iNOS activation, which might contribute to the identification of new targets for iTreg-based treatment in LN (95).

The above shows the role of lncRNA in SLE during the past 2 years. More information on studies over the past 5 years is provided in Table 4.

Table 4
www.frontiersin.org

Table 4. LncRNA alteration in SLE (over the past 5 years).

2.3.3 Circular RNAs in SLE

CircRNAs, which form covalently closed RNA circles, regulate gene expression at both transcriptional and posttranscriptional levels. Acting as miRNA sponges, circRNA can competitively bind mRNA, thereby weakening miRNA-mediated gene suppression (118, 119). Compared with miRNA and lncRNA, circRNA is more stable in mammalian cells, suggesting that circRNAs may be ideal biomarkers for human diseases. The role of circRNAs in SLE has garnered much attention in recent years.

Recently, researchers have used co-expression network analysis, bioinformatics analysis, and multilayer integrative analysis to profile the expression of circRNAs in SLE patients. Circ-calmodulin binding transcription activator 1 (CAMTA1) was found significantly decreased in SLE T cells, and was associated with disease activity. In SLE T cells, upregulated IFN-α inhibited circ-CAMTA1 expression, which may influence glucose metabolism and lead to overexpression of miR-181c-5p, thus decreased the secretion of IL-2 (120). The expression of circ-Rac GTPase activating protein 1 (RACGAP1) was downregulated in SLE PBMCs and related to SLEDAI, anti-dsDNA, and C3 levels, which participated in SLE pathogenesis by regulating the PTEN/AKT signaling pathway through binding to miR-22-3p (121). CircGARS (hsa_circ_0009000) was significantly upregulated in SLE PBMCs, which directly combined with miR-19a to regulate the expression of YTH domain-containing family protein 2 (YTHDF2) and promoted the development of SLE via the A20/NF-κB axis (122). It was also reported that increased expression of hsa_circ_0010957 in SLE CD4+ T cells promoted the secretion of IL-18, IL-6, and IL-17 by mediating the miR−125b/STAT3 signaling pathway, contributing to the pathogenesis of SLE (123).

Additionally, Zheng et al. explored the regulatory mechanisms of circRNAs in SLE patients, revealing a potential relationship between the circRNA–microRNA–mRNA regulatory network and pathogenesis of SLE. They dentified that 131 upregulated and 314 downregulated circRNAs in the plasma of SLE patients, with 28 upregulated and 119 downregulated circRNAs overlapping between PBMCs and plasma, which were enriched in ubiquitination, the TNF signaling pathway and the MAPK pathway. Furthermore, they constructed a network including 54 circRNAs, 41 miRNAs, and 2602 mRNAs to understand the regulatory role of circRNAs in SLE pathogenesis, suggesting that circRNAs in this network could serve as a potential diagnostic biomarker of SLE (124). Zou et al. found that circ-ETS Proto-Oncogene 1 (ETS1) was significantly downregulated in SLE CD4+ T cells, and positively correlated to ANA and anti-dsDNA levels while negatively correlated to C3 levels. After transfection of circETS1 overexpression, CD4+T cells differentiated into Treg cells, causing an imbalance in the Th17/Treg ratio. Transfection of miR-1205 mimic and si-FoxP3 reversed the effects of circETS1 overexpression. In addition, inhibition of miR-1205 had therapeutic effects in SLE mice models. Downregulation of circETS1 promoted SLE activity and inhibited Treg cell differentiation through miR-1205/FoxP3 molecular axis, which may be a novel target for SLE treatment (125).

More information on studies on circRNAs in SLE patients over the past 5 years is provided in Table 5.

Table 5
www.frontiersin.org

Table 5. CircRNA alteration in SLE (over the past 5 years).

2.4 RNA methylation in SLE

In recent years, methylation modifications have been reported to occur not only in DNA, but also occurs in RNA. RNA modifications are post-transcriptional and can alter RNA function. The most common RNA modification is N6-methyladenosine (m6A), which involves methylation of its adenine. M6A methylation is regulated by methyltransferases and demethyltransferases, and it is recognized by RNA binding proteins (142) (Figure 1). Several studies have reported that abnormal m6A modifications can contribute to the pathogenesis of autoimmune diseases by altering the expression of crucial immune factors (143, 144).

Deng et al. found that the expression of AlkB homolog 5 (ALKBH5), a demethyltransferase, was downregulated in both PBMCs and T cells of SLE patients. The expression of ALKBH5 was associated with clinical indicators in SLE, and its downregulation inhibited apoptosis and promoted T cell proliferation, potentially contributing to the development of SLE (145). Liu et al. reported the aberrant m6A methylation in SLE PBMCs. Methyltransferase 3 (METTL3) was upregulated in both SLE and the kidney of MRL/lpr mice, and it promoted the expression of interferon regulatory factor 4 (IRF4), a gene upregulated by m6A. METTL3 induced kidney damage by promoting IRF4-mediated plasma cell infiltration in an m6A-dependent manner (146). Tian et al. identified a causal relationship between MMP9, an m6A-related gene, and ischemic stroke in SLE. As a biomarker for ischemic stroke in SLE, the odds ratio (OR) was 1.0134 (147). Zhao et al. confirmed that the levels of METTL3, Wilms tumor 1 associated protein (WTAP), YTH domain containing 2 (YTHDC2), YTHDF1, fragile X mental retardation 1 (FMR1), and fat mass and obesity-related protein (FTO) in the glomeruli could effectively distinguish patients with LN from healthy individuals, and that they are correlated with the glomerular filtration rate (GFR) and activated natural killer (NK) cells, indicating that they are potential prognostic biomarkers (148).

More classical pathogenic mechanisms of RNA methylation in SLE over the past 5 years are listed in Table 6.

Table 6
www.frontiersin.org

Table 6. Altered RNA methylation genes in SLE (over the past 5 years).

3 Epigenetic factors as potential biomarkers and therapeutic targets for SLE

As currently available biomarkers of SLE, such as autoantibodies, have limitations in early diagnosis, it is crucial to explore new biomarkers that have high specificity and sensitivity for diagnosing the disease and assessing the severity. Increasing evidence suggests that dysregulated epigenetic modifications in immune cells play a significant role in the pathogenesis of SLE (Figure 2). These epigenetic changes have also been identified as potential biomarkers and therapeutic targets.

Figure 2
www.frontiersin.org

Figure 2. Epigenetic regulation of adaptive immune cells in SLE. Adaptive immune cells with altered epigenetic marks in SLE include dendritic cells, T cells and B cells. The figure highlights hypermethylated and hypomethylated genes, histone modification as well as dysregulated noncoding RNAs. CD5-E1B, CD5 protein and exon 1B; HRSE-1, HTLV-1-related endogenous sequence 1; ITGAL, Integrin Subunit Alpha L; MX1, Myxoma resistance 1; NOTCH1, Neurogenic locus notch homolog protein 1; TRIM22, Tripartite Motif Containing 22.

DNA methylation is currently the most studied and stable epigenetic modification. The methylation level of the IFI44L promoter has been suggested as a marker that can diagnose in the early stage and potentially predict specific disease manifestations in SLE (150). Zhang et al. published a study on a high-resolution melting-quantitative polymerase chain reaction assay to detect the methylation of the IFI44L promoter for the diagnosis of SLE, which has good consistency with previous pyrosequencing and is simpler and more economical (151). MicroRNAs are smaller than the transcripts of protein-coding genes, which makes microRNAs more resistant to degradation by endogenous RNase enzymes. Moreover, miRNAs have been found in body fluids such as plasma and urine (152), which makes the test samples easier to obtain. Therefore, miRNAs are important biomarkers for the diagnosis, staging, classification, and prognosis of SLE. In addition to the pathogenesis, miRNA dysregulation is related to disease activity, autoantibody production, organ damage, and therapeutic effects. Chen et al. constructed a multi-miRNA detection platform using target-triggered locked hairpin DNA-functionalized Au nanoprobes for the diagnosis and classification of SLE. This platform achieved an AUC value of 1.00 for a combined urinary sEV miRNA signature (miR-146a, miR-29c, and miR-150). It exhibited good practicability in SLE diagnosis, offering advantages such as low cost, rapidity, high sensitivity, and noninvasiveness (54). Compared with miRNAs, lncRNAs are more tissue-specific and biologically complex (46). Therefore, lncRNA is even more advantageous than miRNA as a new biomarker. Many studies have shown that lncRNAs can be used as potential biomarkers for SLE. For instance, upregulation of H19 promoted apoptosis and the inflammatory response of PBMCs by interacting with miR-19b, contributing to the pathogenesis of SLE. And the AUC value of H19 was 0.853 for SLE diagnosis (94). Additionally, lncNEAT1_2 expression was positively correlated with disease activity in cSLE. According to ROC curve analysis, the AUC value was 0.812, with 62.2% sensitivity and 92.5% specificity, suggesting that lncNEAT1_2 may be a potential biomarker for cSLE (89). Chen et al. constructed a ceRNA network combined with clinical validation to screen for potential molecular markers of SLE through bioinformatics analysis. The results confirmed that the lncRNA XIST/miR-381-3P/STAT1 axis could serve as a molecular marker for SLE diagnosis (76). CircRNAs have become a focus of research in many human diseases. Many studies have revealed that circRNAs can manipulate miRNAs and thus have great potential for clinical applications in diseases such as SLE. There have been many studies demonstrating the circRNA expression profile in SLE through various technologies. Through these analyses, researchers have proved that many circRNAs have characteristics for the prediction of disease progression. A circRNA–microRNA–mRNA regulatory network including 54 circRNAs, 41 miRNAs, and 2602 mRNAs was constructed to understand the regulatory role of circRNAs in SLE pathogenesis, which could be potential diagnostic biomarkers of SLE (124).

Epigenetic modifications can be used not only as potential biomarkers, but also as potential therapeutic targets for lupus. In the SLE mouse model, miR-155 inhibition improved Treg function under inflammatory stimulation and alleviated SLE (56). After treatment with UC-MSCs modified by miR-125b-5p in MRL/lpr mice, and inflammatory infiltration and microthrombus formation in the lungs and kidneys were reduced (57). In addition, downregulation of circETS1 promoted SLE activity and inhibited Treg cell differentiation through miR-1205/FoxP3 molecular axis, which may be a novel target for SLE treatment (125).

More potential epigenetic biomarkers and potential therapeutic targets for SLE over the past 5 years are listed in Tables 7 and 8.

Table 7
www.frontiersin.org

Table 7. Potential epigenetic biomarkers for SLE (over the past 5 years).

Table 8
www.frontiersin.org

Table 8. Potential therapeutic targets for SLE (over the past 5 years).

4 Conclusions

Discoveries of epigenetic modifications have extended our knowledge of complex regulation in genes and added new insights into understanding the pathogenesis of SLE. There are still challenges in ascertaining the mechanism of epigenetic changes in the occurrence and development of the disease. Epigenetic events have great potential in finding targets for individualized treatment interventions and disease diagnosis biomarkers. With the increasing attention to epigenetic mechanisms, more surprising discoveries will provide new ideas for the treatment of SLE.

Author contributions

XZ: Writing – original draft, Writing – review & editing. SZ: Writing – original draft, Writing – review & editing. YL: Conceptualization, Funding acquisition, Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by the Natural Science Foundation of Changsha, Hunan, China (NO: kq2208323).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Moulton VR, Suarez-Fueyo A, Meidan E, Li H, Mizui M, Tsokos GC. Pathogenesis of human systemic lupus erythematosus: A cellular perspective. Trends Mol Med. (2017) 23:615–35. doi: 10.1016/j.molmed.2017.05.006

PubMed Abstract | Crossref Full Text | Google Scholar

2. Harley JB, Kelly JA, Kaufman KM. Unraveling the genetics of systemic lupus erythematosus. Springer Semin Immunopathol. (2006) 28:119–30. doi: 10.1007/s00281-006-0040-5

PubMed Abstract | Crossref Full Text | Google Scholar

3. Rhodes B, Vyse TJ. The genetics of SLE: an update in the light of genome-wide association studies. Rheumatol (Oxford). (2008) 47:1603–11. doi: 10.1093/rheumatology/ken247

PubMed Abstract | Crossref Full Text | Google Scholar

4. Cooper GS, Parks CG, Treadwell EL, St Clair EW, Gilkeson GS, Dooley MA. Occupational risk factors for the development of systemic lupus erythematosus. J Rheumatol. (2004) 31:1928–33.

Google Scholar

5. Aslani S, Mahmoudi M, Karami J, Jamshidi AR, Malekshahi Z, Nicknam MH. Epigenetic alterations underlying autoimmune diseases. Autoimmunity. (2016) 49:69–83. doi: 10.3109/08916934.2015.1134511

PubMed Abstract | Crossref Full Text | Google Scholar

6. Richardson B. Effect of an inhibitor of DNA methylation on T cells. II. 5-Azacytidine induces self-reactivity in antigen-specific T4+ cells. Hum Immunol. (1986) 17:456–70. doi: 10.1016/0198-8859(86)90304-6

PubMed Abstract | Crossref Full Text | Google Scholar

7. Liang P, Song F, Ghosh S, Morien E, Qin M, Mahmood S, et al. Genome-wide survey reveals dynamic widespread tissue-specific changes in DNA methylation during development. BMC Genomics. (2011) 12:231. doi: 10.1186/1471-2164-12-231

PubMed Abstract | Crossref Full Text | Google Scholar

8. Zhao M, Wang J, Liao W, Li D, Li M, Wu H, et al. Increased 5-hydroxymethylcytosine in CD4(+) T cells in systemic lupus erythematosus. J Autoimmun. (2016) 69:64–73. doi: 10.1016/j.jaut.2016.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

9. WYATT GR, COHEN SS. A new pyrimidine base from bacteriophage nucleic acids. Nature. (1952) 170:1702–1702. doi: 10.1038/1701072a0

PubMed Abstract | Crossref Full Text | Google Scholar

10. Liang J, Yang F, Zhao L, Bi C, Cai B. Physiological and pathological implications of 5-hydroxymethylcytosine in diseases. Oncotarget. (2016) 7:48813–31. doi: 10.18632/oncotarget.v7i30

PubMed Abstract | Crossref Full Text | Google Scholar

11. Tan Li, Shi. YG. Tet family proteins and 5-hydroxymethylcytosine in development and disease. Development. (2012) 139:1895–902. doi: 10.1242/dev.070771

PubMed Abstract | Crossref Full Text | Google Scholar

12. Li Y, Zhao M, Yin H, Gao F, Wu X, Luo Y, et al. Overexpression of the growth arrest and DNA damage-induced 45alpha gene contributes to autoimmunity by promoting DNA demethylation in lupus T cells. Arthritis Rheum. (2010) 62:1438–47. doi: 10.1002/art.27363

PubMed Abstract | Crossref Full Text | Google Scholar

13. Wu H, Zhao M, Tan L, Lu Q. The key culprit in the pathogenesis of systemic lupus erythematosus: Aberrant DNA methylation. Autoimmun Rev. (2016) 15:684–9. doi: 10.1016/j.autrev.2016.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

14. Lu Q, Wu A, Richardson BC. Demethylation of the same promoter sequence increases CD70 expression in lupus T cells and T cells treated with lupus-inducing drugs. J Immunol. (2005) 174:6212–9. doi: 10.4049/jimmunol.174.10.6212

PubMed Abstract | Crossref Full Text | Google Scholar

15. Lu Q, Kaplan M, Ray D, Ray D, Zacharek S, Gutsch D, et al. Demethylation of ITGAL (CD11a) regulatory sequences in systemic lupus erythematosus. Arthritis Rheum. (2002) 46:1282–91. doi: 10.1002/art.10234

PubMed Abstract | Crossref Full Text | Google Scholar

16. Lu Q, Wu A, Tesmer L, Ray D, Yousif N, Richardson B. Demethylation of CD40LG on the inactive X in T cells from women with lupus. J Immunol. (2007) 179:6352–8. doi: 10.4049/jimmunol.179.9.6352

PubMed Abstract | Crossref Full Text | Google Scholar

17. Kaplan MJ, Lu Q, Wu A, Attwood J, Richardson B. Demethylation of promoter regulatory elements contributes to perforin overexpression in CD4+ lupus T cells. J Immunol. (2004) 172:3652–61. doi: 10.4049/jimmunol.172.6.3652

PubMed Abstract | Crossref Full Text | Google Scholar

18. Liao L, Li S, Upreti B, Wang X, Yang Y, Lou X, et al. Status of TWEAK DNA methylation and mRNA expression in systemic lupus erythematosus. Lupus. (2023) 32:171–9. doi: 10.1177/09612033221141261

PubMed Abstract | Crossref Full Text | Google Scholar

19. Dong Z, Zhang B, Rong J, Yang X, Wang Y, Zhang Q, et al. The aberrant expression of CD45 isoforms and levels of sex hormones in systemic lupus erythematosus. Clin Rheumatol. (2022) 41:1087–93. doi: 10.1007/s10067-021-05934-x

PubMed Abstract | Crossref Full Text | Google Scholar

20. Gao B. Identification of feature autophagy-related genes and DNA methylation profiles in systemic lupus erythematosus patients. Med Sci Monit. (2021) 27:e933425. doi: 10.12659/MSM.933425

PubMed Abstract | Crossref Full Text | Google Scholar

21. Luo S, Wu R, Li Q, Zhang G. Epigenetic regulation of IFI44L expression in monocytes affects the functions of monocyte-derived dendritic cells in systemic lupus erythematosus. J Immunol Res. (2022) 2022:4053038. doi: 10.1155/2022/4053038

PubMed Abstract | Crossref Full Text | Google Scholar

22. Hurtado C, Rojas-Gualdrón DF, Urrego R, Cashman K, Vásquez-Trespalacios EM, Díaz-Coronado JC, et al. Altered B cell phenotype and CD27+ memory B cells are associated with clinical features and environmental exposure in Colombian systemic lupus erythematosus patients. Front Med (Lausanne). (2022) 9:950452. doi: 10.3389/fmed.2022.950452

PubMed Abstract | Crossref Full Text | Google Scholar

23. Kurata I, Mikami N, Ohyama A, Osada A, Kondo Y, Tsuboi H, et al. Impaired function of PD-1(+) follicular regulatory T cells in systemic lupus erythematosus. Clin Exp Immunol. (2021) 206:28–35. doi: 10.1111/cei.13643

PubMed Abstract | Crossref Full Text | Google Scholar

24. Liu L, Hu L, Yang L, Jia S, Du P, Min X, et al. UHRF1 downregulation promotes T follicular helper cell differentiation by increasing BCL6 expression in SLE. Clin Epigenet. (2021) 13:31. doi: 10.1186/s13148-021-01007-7

PubMed Abstract | Crossref Full Text | Google Scholar

25. Ehtesham N, Mosallaei M, Zaboli Mahdiabadi M, Kenarangi T, Farhadi A, Heidari MF, et al. Significant hypomethylation of MMP9 gene promoter in patients with systemic lupus erythematosus. Lupus. (2023) 32:1056–65. doi: 10.1177/09612033231185618

PubMed Abstract | Crossref Full Text | Google Scholar

26. Ehtesham N, Alesaeidi S, Mohammad Zadeh D, Saghaei M, Fakhri M, Bayati Z, et al. Significant heightened methylation levels of RUNX3 gene promoter in patients with systemic lupus erythematosus. Lupus. (2024) 33:547–54. doi: 10.1177/09612033241241850

PubMed Abstract | Crossref Full Text | Google Scholar

27. Liu X, Zhou S, Huang M, Zhao M, Zhang W, Liu Q, et al. DNA methylation and whole-genome transcription analysis in CD4(+) T cells from systemic lupus erythematosus patients with or without renal damage. Clin Epigenet. (2024) 16:98. doi: 10.1186/s13148-024-01699-7

PubMed Abstract | Crossref Full Text | Google Scholar

28. Li Y, Zhang S, Tang C, Yang B, Atrooz F, Ren Z, et al. Autoimmune and neuropsychiatric phenotypes in a Mecp2 transgenic mouse model on C57BL/6 background. Front Immunol. (2024) 15:1370254. doi: 10.3389/fimmu.2024.1370254

PubMed Abstract | Crossref Full Text | Google Scholar

29. Ugarte-Berzal E, Boon L, Martens E, Rybakin V, Blockmans D, Vandooren J, et al. MMP-9/gelatinase B degrades immune complexes in systemic lupus erythematosus. Front Immunol. (2019) 10:538. doi: 10.3389/fimmu.2019.00538

PubMed Abstract | Crossref Full Text | Google Scholar

30. Koga T, Ichinose K, Kawakami A, Tsokos GC. The role of IL-17 in systemic lupus erythematosus and its potential as a therapeutic target. Expert Rev Clin Immunol. (2019) 15:629–37. doi: 10.1080/1744666X.2019.1593141

PubMed Abstract | Crossref Full Text | Google Scholar

31. Zhang C, Zhang C, Ji J, Xiong X, Lu Y. Hsa_circ_0012919 regulates expression of MDA5 by miR-125a-3p in CD4+ T cells of systemic lupus erythematous. Lupus. (2020) 29:727–34. doi: 10.1177/0961203320920706

PubMed Abstract | Crossref Full Text | Google Scholar

32. Wardowska A, Komorniczak M, Bułło-Piontecka B, Dȩbska-Ślizień MA, Pikuła M. Transcriptomic and epigenetic alterations in dendritic cells correspond with chronic kidney disease in lupus nephritis. Front Immunol. (2019) 10:2026. doi: 10.3389/fimmu.2019.02026

PubMed Abstract | Crossref Full Text | Google Scholar

33. Rice JC, Allis CD. Histone methylation versus histone acetylation: new insights into epigenetic regulation. Curr Opin Cell Biol. (2001) 13:263–73. doi: 10.1016/S0955-0674(00)00208-8

PubMed Abstract | Crossref Full Text | Google Scholar

34. Singh SK, Bahal R, Rasmussen TP. Evidence that miR-152-3p is a positive regulator of SETDB1-mediated H3K9 histone methylation and serves as a toggle between histone and DNA methylation. Exp Cell Res. (2020) 395:112216. doi: 10.1016/j.yexcr.2020.112216

PubMed Abstract | Crossref Full Text | Google Scholar

35. Zhang L, Tian S, Zhao M, Yang T, Quan S, Yang Q, et al. SUV39H1-DNMT3A-mediated epigenetic regulation of Tim-3 and galectin-9 in the cervical cancer. Cancer Cell Int. (2020) 20:325. doi: 10.1186/s12935-020-01380-y

PubMed Abstract | Crossref Full Text | Google Scholar

36. Gautam P, Sharma A, Bhatnagar A. Global histone modification analysis reveals hypoacetylated H3 and H4 histones in B Cells from systemic lupus erythematosus patients. Immunol Lett. (2021) 240:41–5. doi: 10.1016/j.imlet.2021.09.007

PubMed Abstract | Crossref Full Text | Google Scholar

37. Zhang Q, Liu Y, Liao J, Wu R, Zhan Y, Zhang P, et al. Deficiency of p53 Causes the Inadequate Expression of miR-1246 in B Cells of Systemic Lupus Erythematosus. J Immunol. (2022) 209:1492–8. doi: 10.4049/jimmunol.2200307

PubMed Abstract | Crossref Full Text | Google Scholar

38. Luo S, Zhang H, Xie Y, Huang J, Luo D, Zhang Q. Decreased SUV39H1 at the promoter region leads to increased CREMα and accelerates autoimmune response in CD4(+) T cells from patients with systemic lupus erythematosus. Clin Epigenet. (2022) 14:181. doi: 10.1186/s13148-022-01411-7

PubMed Abstract | Crossref Full Text | Google Scholar

39. Luo S, Xie Y, Huang J, Liu J, Zhang Q. Decreased jumonji domain-containing 3 at the promoter downregulates hematopoietic progenitor kinase 1 expression and cytoactivity of T follicular helper cells from systemic lupus erythematosus patients. J Immunol Res. (2022) 2022:3690892. doi: 10.1155/2022/3690892

PubMed Abstract | Crossref Full Text | Google Scholar

40. Liu Y, Luo S, Zhan Y, Wang J, Zhao R, Li Y, et al. Increased expression of PPAR-γ Modulates monocytes into a M2-like phenotype in SLE patients: an implicative protective mechanism and potential therapeutic strategy of systemic lupus erythematosus. Front Immunol. (2020) 11:579372. doi: 10.3389/fimmu.2020.579372

PubMed Abstract | Crossref Full Text | Google Scholar

41. Fan J, Iwata S, Tanaka Y, Kitanaga Y, Ishii A, Maiko H, et al. Kdm5a promotes B cell activation in systemic lupus erythematosus via downregulation of A20 by histone modification. Pathol Res Pract. (2021) 9:153653. doi: 10.1016/j.prp.2021.153653

PubMed Abstract | Crossref Full Text | Google Scholar

42. Ding S, Zhang Q, Luo S, Gao L, Huang J, Lu J, et al. BCL-6 suppresses miR-142-3p/5p expression in SLE CD4(+) T cells by modulating histone methylation and acetylation of the miR-142 promoter. Cell Mol Immunol. (2020) 17:474–82. doi: 10.1038/s41423-019-0268-3

PubMed Abstract | Crossref Full Text | Google Scholar

43. Lee S, Nakayamada S, Kubo S, Yamagata K, Yoshinari H, Tanaka Y. Interleukin-23 drives expansion of Thelper 17 cells through epigenetic regulation by signal transducer and activators of transcription 3 in lupus patients. Rheumatol (Oxford). (2020) 59:3058–69. doi: 10.1093/rheumatology/keaa176

PubMed Abstract | Crossref Full Text | Google Scholar

44. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. (2012) 489:57–74. doi: 10.1038/nature11247

PubMed Abstract | Crossref Full Text | Google Scholar

45. Eulalio A, Huntzinger E, Izaurralde E. Getting to the root of miRNA-mediated gene silencing. Cell. (2008) 132:9–14. doi: 10.1016/j.cell.2007.12.024

PubMed Abstract | Crossref Full Text | Google Scholar

46. Wu GC, Li J, Leng RX, Li XP, Li XM, Wang DG, et al. Identification of long non-coding RNAs GAS5, linc0597 and lnc-DC in plasma as novel biomarkers for systemic lupus erythematosus. Oncotarget. (2017) 8:23650–63. doi: 10.18632/oncotarget.15569

PubMed Abstract | Crossref Full Text | Google Scholar

47. Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet. (2008) 9:102–14. doi: 10.1038/nrg2290

PubMed Abstract | Crossref Full Text | Google Scholar

48. Bushati N, Cohen SM. microRNA functions. Annu Rev Cell Dev Biol. (2007) 23:175–205. doi: 10.1146/annurev.cellbio.23.090506.123406

PubMed Abstract | Crossref Full Text | Google Scholar

49. Luo S, Wu R, Li Q, Zhang G. MiR-301a-3p advances IRAK1-mediated differentiation of th17 cells to promote the progression of systemic lupus erythematosus via targeting PELI1. J Healthc Eng. (2021) 2021:2982924. doi: 10.1155/2021/2982924

PubMed Abstract | Crossref Full Text | Google Scholar

50. Yang M, Yang B, Deng D. Targeting of EIF4EBP1 by miR-99a-3p affects the functions of B lymphocytes via autophagy and aggravates SLE disease progression. J Cell Mol Med. (2021) 25:10291–305. doi: 10.1111/jcmm.v25.21

PubMed Abstract | Crossref Full Text | Google Scholar

51. Chen J, Peng L, Zhao Z, Yang Q, Yin F, Liu M, et al. HDAC1 potentiates CD4 + T cell activation by inhibiting miR-124 and promoting IRF1 in systemic lupus erythematosus. Cell Immunol. (2021) 362:104284. doi: 10.1016/j.cellimm.2021.104284

PubMed Abstract | Crossref Full Text | Google Scholar

52. Tao B, Xiang W, Li X, He C, Chen L, Xia X, et al. Regulation of Toll-like receptor-mediated inflammatory response by microRNA-152-3p-mediated demethylation of MyD88 in systemic lupus erythematosus. Inflammation Res. (2021) 70:285–96. doi: 10.1007/s00011-020-01433-y

PubMed Abstract | Crossref Full Text | Google Scholar

53. Wu YH, Kuo CF, Hsieh AH, Hsieh HL, Chan YF, Hwang TL. Upregulation of miR-210-5p impairs dead cell clearance by macrophages through the inhibition of Sp1-and HSCARG-dependent NADPH oxidase pathway. Free Radic Biol Med. (2021) 172:441–50. doi: 10.1016/j.freeradbiomed.2021.06.029

PubMed Abstract | Crossref Full Text | Google Scholar

54. Chen X, Xiang Q, Yan S, Wang Y, Su N, Yang X, et al. Simultaneous multi-miRNA detection in urinary small extracellular vesicles using target-triggered locked hairpin DNA-functionalized au nanoprobes for systemic lupus erythematosus diagnosis. Anal Chem. (2024) 96:16370–8. doi: 10.1021/acs.analchem.4c03794

PubMed Abstract | Crossref Full Text | Google Scholar

55. ElFeky DS, Omar NM, Shaker OG, Abdelrahman W, Gheita TA, Nada MG. Circulatory microRNAs and proinflammatory cytokines as predictors of lupus nephritis. Front Immunol. (2024) 15:1449296. doi: 10.3389/fimmu.2024.1449296

PubMed Abstract | Crossref Full Text | Google Scholar

56. Yu J, Mei J, Zuo D, Zhang M, Yu S, Li F, et al. Inflammatory factor-mediated miR-155/SOCS1 signaling axis leads to Treg impairment in systemic lupus erythematosus. Int Immunopharmacol. (2024) 141:113013. doi: 10.1016/j.intimp.2024.113013

PubMed Abstract | Crossref Full Text | Google Scholar

57. Wu Z, Hu M, Zhao Q, Lv F, Zhang J, Zhang W, et al. Immunomodulatory mechanism of umbilical cord mesenchymal stem cells modified by miR-125b-5p in systemic lupus erythematosus. Beijing Da Xue Xue Bao Yi Xue Ban. (2024) 56:860–7. doi: 10.19723/j.issn.1671-167X.2024.05.017

PubMed Abstract | Crossref Full Text | Google Scholar

58. Zhou S, Zhang J, Luan P, Ma Z, Dang J, Zhu H, et al. miR-183-5p is a potential molecular marker of systemic lupus erythematosus. J Immunol Res. (2021) 2021:5547635. doi: 10.1155/2021/5547635

PubMed Abstract | Crossref Full Text | Google Scholar

59. Zhao X, Li S, Wang Z, Bai N, Feng Y. miR−101−3p negatively regulates inflammation in systemic lupus erythematosus via MAPK1 targeting and inhibition of the NF−κB pathway. Mol Med Rep. (2021) 23:359. doi: 10.3892/mmr.2021.11998

PubMed Abstract | Crossref Full Text | Google Scholar

60. Sun H, Guo F, Xu L. Downregulation of microRNA-101-3p participates in systemic lupus erythematosus progression via negatively regulating HDAC9. J Cell Biochem. (2020) 121:4310–20. doi: 10.1002/jcb.v121.10

PubMed Abstract | Crossref Full Text | Google Scholar

61. Yang X, Shi L, Zheng X, Liu X, Qian J. Modulation of miR-548m encoded by X chromosome on the PTEN pathway in systemic lupus erythematosus. Clin Exp Rheumatol. (2022) 40:56–63. doi: 10.55563/clinexprheumatol/yjsbqm

PubMed Abstract | Crossref Full Text | Google Scholar

62. Yuan S, Tang C, Chen D, Li F, Huang M, Ye J, et al. miR-98 modulates cytokine production from human PBMCs in systemic lupus erythematosus by targeting IL-6 mRNA. J Immunol Res. (2019) 2019:9827574. doi: 10.1155/2019/9827574

PubMed Abstract | Crossref Full Text | Google Scholar

63. Shi X, Ye L, Xu S, Guo G, Zuo Z, Ye M, et al. Downregulated miR−29a promotes B cell overactivation by upregulating Crk−like protein in systemic lupus erythematosus. Mol Med Rep. (2020) 22:841–9. doi: 10.3892/mmr.2020.11166

PubMed Abstract | Crossref Full Text | Google Scholar

64. Luo S, Ding S, Liao J, Zhang P, Liu Y, Zhao M, et al. Excessive miR-152-3p results in increased BAFF expression in SLE B-cells by inhibiting the KLF5 expression. Front Immunol. (2019) 10:1127. doi: 10.3389/fimmu.2019.01127

PubMed Abstract | Crossref Full Text | Google Scholar

65. Xia Y, Tao JH, Fang X, Xiang N, Dai XJ, Jin L, et al. MicroRNA-326 upregulates B cell activity and autoantibody production in lupus disease of MRL/lpr mice. Mol Ther Nucleic Acids. (2018) 11:284–91. doi: 10.1016/j.omtn.2018.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

66. Gong A, Mi L, Wei F, Zhuang Y, Song Y, Pan C, et al. Downregulation of miR-137 Facilitates CD4+ T Cell Pyroptosis in Systemic Lupus Erythematosus via Stimulating AMPK Pathway. J Immunol Res. (2023) 2023:1241774. doi: 10.1155/2023/1241774

PubMed Abstract | Crossref Full Text | Google Scholar

67. Wang H, Geng G, Zhang D, Han F, Ye S. Analysis of microRNA-199a-3p expression in CD4(+) T cells of systemic lupus erythematosus. Clin Rheumatol. (2023) 42:1683–94. doi: 10.1007/s10067-023-06534-7

PubMed Abstract | Crossref Full Text | Google Scholar

68. Latini A, Ciccacci C, Benedittis G, Novelli L, Ceccarelli F, Conti F, et al. Altered expression of miR-142, miR-155, miR-499a and of their putative common target MDM2 in systemic lupus erythematosus. Epigenomics. (2021) 13:5–13. doi: 10.2217/epi-2020-0278

PubMed Abstract | Crossref Full Text | Google Scholar

69. Hiramatsu-Asano S, Sunahori-Watanabe K, Zeggar S, Katsuyama E, Mukai T, Morita Y, et al. Deletion of mir223 exacerbates lupus nephritis by targeting S1pr1 in fas(lpr/lpr) mice. Front Immunol. (2020) 11:616141. doi: 10.3389/fimmu.2020.616141

PubMed Abstract | Crossref Full Text | Google Scholar

70. Suo QF, Sheng J, Qiang FY, Tang ZS, Yang YY. Association of long non-coding RNA GAS5 and miR-21 levels in CD4(+) T cells with clinical features of systemic lupus erythematosus. Exp Ther Med. (2018) 15:345–50. doi: 10.3892/etm.2017.5429

PubMed Abstract | Crossref Full Text | Google Scholar

71. Qin H, Chen S, Liu X, Liang J, Wu H, Zhu X. miR-132-3p downregulates FOXO1 in CD4(+) T cells and is associated with disease manifestations in patients with lupus. J Int Med Res. (2024) 52:3000605241286762. doi: 10.1177/03000605241286762

PubMed Abstract | Crossref Full Text | Google Scholar

72. Yang B, Huang X, Xu S, Li L, Wu W, Dai Y, et al. Decreased miR-4512 levels in monocytes and macrophages of individuals with systemic lupus erythematosus contribute to innate immune activation and neutrsophil NETosis by targeting TLR4 and CXCL2. Front Immunol. (2021) 12:756825. doi: 10.3389/fimmu.2021.756825

PubMed Abstract | Crossref Full Text | Google Scholar

73. Teng Z, Lin X, Luan C, Sun Y, Li X. The high expression of miR-564 in patients with systemic lupus erythematosus promotes differentiation and maturation of DC cells by negatively regulating TP53 expression in vitro. Lupus. (2021) 30:1469–80. doi: 10.1177/09612033211020367

PubMed Abstract | Crossref Full Text | Google Scholar

74. Mishra R, Bhattacharya S, Rawat BS, Kumar A, Kumar A, Niraj K, et al. MicroRNA-30e-5p has an Integrated Role in the Regulation of the Innate Immune Response during Virus Infection and Systemic Lupus Erythematosus. iScience. (2020) 23:101322. doi: 10.1016/j.isci.2020.101322

PubMed Abstract | Crossref Full Text | Google Scholar

75. Zhang L, Zhang X, Si F. MicroRNA-124 represents a novel diagnostic marker in human lupus nephritis and plays an inhibitory effect on the growth and inflammation of renal mesangial cells by targeting TRAF6. Int J Clin And Exp Pathol. (2019) 12:1578–88.

PubMed Abstract | Google Scholar

76. Chen J, Li M, Shang S, Cheng L, Tang Z, Huang C. LncRNA XIST/miR-381-3P/STAT1 axis as a potential biomarker for lupus nephritis. Lupus. (2024) 33:1176–91. doi: 10.1177/09612033241273072

PubMed Abstract | Crossref Full Text | Google Scholar

77. Zhang H, Zhou X, Li Q, Zheng M. Clinical significance of miR-200a in systemic lupus erythematosus and renal damage in children. Cell Mol Biol (Noisy-le-grand). (2024) 70:94–8. doi: 10.14715/cmb/2024.70.1.13

PubMed Abstract | Crossref Full Text | Google Scholar

78. Nakhjavani M, Etemadi J, Pourlak T, Mirhosaini Z, Zununi Vahed S, Abediazar S. Plasma levels of miR-21, miR-150, miR-423 in patients with lupus nephritis. Iran J Kidney Dis. (2019) 13:198–206.

PubMed Abstract | Google Scholar

79. Wu L, Han X, Jiang X, Ding H, Qi C, Yin Z, et al. Downregulation of renal hsa-miR-127-3p contributes to the overactivation of type I interferon signaling pathway in the kidney of lupus nephritis. Front Immunol. (2021) 12:747616. doi: 10.3389/fimmu.2021.747616

PubMed Abstract | Crossref Full Text | Google Scholar

80. Li X, Luo F, Li J, Luo C. MiR-183 delivery attenuates murine lupus nephritis-related injuries via targeting mTOR. Scandinavian J Immunol. (2019) 90:e12810. doi: 10.1111/sji.12810

PubMed Abstract | Crossref Full Text | Google Scholar

81. Qi H, Cao Q, Liu Q. MicroRNA-183 exerts a protective role in lupus nephritis through blunting the activation of TGF-β/Smad/TLR3 pathway via reducing Tgfbr1. Exp Cell Res. (2020) 394:112138. doi: 10.1016/j.yexcr.2020.112138

PubMed Abstract | Crossref Full Text | Google Scholar

82. Zheng J, Guo R, Tang Y, Fu Q, Chen J, Wu L, et al. miR-152 attenuates the severity of lupus nephritis through the downregulation of macrophage migration inhibitory factor (MIF)-induced expression of COL1A1. Front Immunol. (2019) 10:158. doi: 10.3389/fimmu.2019.00158

PubMed Abstract | Crossref Full Text | Google Scholar

83. Zhao CN, Mao YM, Liu LN, Li XM, Wang DG, Pan HF. Emerging role of lncRNAs in systemic lupus erythematosus. Biomedicine Pharmacotherapy. (2018) 106:584–92. doi: 10.1016/j.biopha.2018.06.175

PubMed Abstract | Crossref Full Text | Google Scholar

84. Wapinski O, Chang HY. Long noncoding RNAs and human disease. Trends Cell Biol. (2011) 21:354–61. doi: 10.1016/j.tcb.2011.04.001

PubMed Abstract | Crossref Full Text | Google Scholar

85. Zhang Q, Chen CY, Yedavalli VS, Jeang KT. NEAT1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. mBio. (2013) 4:e00596–00512. doi: 10.1128/mBio.00596-12

PubMed Abstract | Crossref Full Text | Google Scholar

86. Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. (2019) 10:1495. doi: 10.1038/s41467-019-09482-6

PubMed Abstract | Crossref Full Text | Google Scholar

87. Xiang M, Wang Y, Chen Q, Wang J, Gao Z, Liang J, et al. LncRNA NEAT1 promotes IL-6 secretion in monocyte-derived dendritic cells via sponging miR-365a-3p in systemic lupus erythematosus. Epigenetics. (2023) 18:2226492. doi: 10.1080/15592294.2023.2226492

PubMed Abstract | Crossref Full Text | Google Scholar

88. Jiang Y, Zhao Y, Mo X. Expression of lncRNA NEAT1 in peripheral blood mononuclear cells of patients with systemic lupus erythematosus and its correlation with Th1/Th2 balance. Int J Clin Exp Pathol. (2021) 14:646–52.

PubMed Abstract | Google Scholar

89. Li S, Wang X, Zhao X, Deng J, Kuang W, Zhang J, et al. Long noncoding nuclear enriched abundant transcript 1_2 is a promising biomarker for childhood-onset systemic lupus erythematosus. Pediatr Investig. (2024) 8:101–7. doi: 10.1002/ped4.12413

PubMed Abstract | Crossref Full Text | Google Scholar

90. Xiao J, Wang D. LncRNA GAS5 as an inflammatory regulator acting through pathway in human lupus. Curr Pharm Des. (2023) 29:1293–9. doi: 10.2174/1381612829666230517102205

PubMed Abstract | Crossref Full Text | Google Scholar

91. Liu CH, Lu YL, Huang HT, Wang CF, Luo HC, Wei GJ, et al. Association of LncRNA-GAS5 gene polymorphisms and PBMC LncRNA-GAS5 level with risk of systemic lupus erythematosus in Chinese population. J Cell Mol Med. (2021) 25:3548–59. doi: 10.1111/jcmm.16438

PubMed Abstract | Crossref Full Text | Google Scholar

92. Liu L, Hu L, Long H, Zheng M, Hu Z, He Y, et al. LncRNA IL21-AS1 interacts with hnRNPU protein to promote IL21 overexpression and aberrant differentiation of Tfh cells in systemic lupus erythematosus. Clin Transl Med. (2022) 12:e1117. doi: 10.1002/ctm2.v12.12

PubMed Abstract | Crossref Full Text | Google Scholar

93. Hao H, Nakayamada S, Ohkubo N, Yamagata K, Zhang M, Shan Y, et al. Involvement of lncRNA IL21-AS1 in interleukin-2 and T follicular regulatory cell activation in systemic lupus erythematosus. Arthritis Res Ther. (2021) 23:302. doi: 10.1186/s13075-021-02682-w

PubMed Abstract | Crossref Full Text | Google Scholar

94. Yang H, Wang S, Wang F, Bai X, Ren J. Serum long non-coding Ribonucleic Acid H19 serves as a biomarker for systemic lupus erythematosus and participates in the disease progression. Lupus. (2024) 33:675–84. doi: 10.1177/09612033241243175

PubMed Abstract | Crossref Full Text | Google Scholar

95. Liu Y, Li P, Wang L, Jiang L, Li Z, Mei Y, et al. iTreg cells-secreted IL10 alleviates lupus nephritis through inactivating lncRNA HAR1A transcription to suppress SMARCD1-mediated iNOS activation. Autoimmunity. (2024) 57:2423758. doi: 10.1080/08916934.2024.2423758

PubMed Abstract | Crossref Full Text | Google Scholar

96. Huang S, Dong D, Zhang Y, Chen Z, Geng J, Zhao Y. Long non-coding RNA nuclear paraspeckle assembly transcript 1 promotes activation of T helper 2 cells via inhibiting STAT6 ubiquitination. Hum Cell. (2021) 34:800–7. doi: 10.1007/s13577-021-00496-1

PubMed Abstract | Crossref Full Text | Google Scholar

97. Dong G, Yang Y, Li X, Yao X, Zhu Y, Zhang H, et al. Granulocytic myeloid-derived suppressor cells contribute to IFN-I signaling activation of B cells and disease progression through the lncRNA NEAT1-BAFF axis in systemic lupus erythematosus. Biochim Biophys Acta Mol Basis Dis. (2020) 1866:165554. doi: 10.1016/j.bbadis.2019.165554

PubMed Abstract | Crossref Full Text | Google Scholar

98. Liu Q, Deng Y, Li C, Xie H, Liu Q, Ming S, et al. LncRNA GAS5 suppresses CD4(+) T cell activation by upregulating E4BP4 via inhibiting miR-92a-3p in systemic lupus erythematosus. Immunol Lett. (2020) 227:41–7. doi: 10.1016/j.imlet.2020.08.001

PubMed Abstract | Crossref Full Text | Google Scholar

99. Wu GC, Hu Y, Guan SY, Ye DQ, Pan HF. Differential plasma expression profiles of long non-coding RNAs reveal potential biomarkers for systemic lupus erythematosus. Biomolecules. (2019) 9:206. doi: 10.3390/biom9060206

PubMed Abstract | Crossref Full Text | Google Scholar

100. Ma Q, Li L, Xing Y. LncRNA NRIR serves as a biomarker for systemic lupus erythematosus and participates in the disease progression. Lupus. (2024) 33(14):1538–46. doi: 10.1177/09612033241294032

PubMed Abstract | Crossref Full Text | Google Scholar

101. Rasuli E, Javidi-Aghdam K, Akbarzadeh-Khiavi M, Abdshah A, Gadakchi L, Jafarpour M, et al. Immunoregulatory role of AC007278.3 and HOTAIR long non-coding RNAs in lupus nephritis: potential biomarkers and therapeutic targets. Mol Biol Rep. (2024) 51:1075. doi: 10.1007/s11033-024-10019-4

PubMed Abstract | Crossref Full Text | Google Scholar

102. Wang C, Yuan S, Zeng Y, Li W, Ye J, Li F, et al. A novel long noncoding RNA ENST00000597482 serves as a potential biomarker for disease activity and diagnosis of systemic lupus erythematosus. Lupus. (2024) 33:1089–99. doi: 10.1177/09612033241266988

PubMed Abstract | Crossref Full Text | Google Scholar

103. Peng XC, Ma LL, Miao JY, Xu SQ, Shuai ZW. Differential lncRNA profiles of blood plasma-derived exosomes from systemic lupus erythematosus. Gene. (2024) 927:148713. doi: 10.1016/j.gene.2024.148713

PubMed Abstract | Crossref Full Text | Google Scholar

104. Jiang L, Qi A, Yang H, Wang S, Wang F, Bai X, et al. LncRNA SNHG1 serves as a biomarker for systemic lupus erythematosus and participates in the disease progression. Apmis. (2024) 132:507–14. doi: 10.1111/apm.v132.7

PubMed Abstract | Crossref Full Text | Google Scholar

105. Zhu X, Chen Y, Yin Z, Zhang Y, Shen Y, Dai D, et al. Novel potential lncRNA biomarker in B cells indicates essential pathogenic pathway activation in patients with SLE. Lupus Sci Med. (2024) 11:e001065. doi: 10.1136/lupus-2023-001065

PubMed Abstract | Crossref Full Text | Google Scholar

106. Alrefai AA, Abouelenin MAH, Salman MMA, Tawfeek GAE, Abbas MA. Expression profile of long-noncoding RNAs MIR31HG, NKILA, and PACER in systemic lupus erythematosus patients. Clin Biochem. (2024) 126:110734. doi: 10.1016/j.clinbiochem.2024.110734

PubMed Abstract | Crossref Full Text | Google Scholar

107. Wang Y, He J, Ma H, Liu J, Du L, Chai C, et al. NR_103776.1 as a novel diagnostic biomarker for systemic lupus erythematosus. Ir J Med Sci. (2024) 193:211–21. doi: 10.1007/s11845-023-03420-8

PubMed Abstract | Crossref Full Text | Google Scholar

108. Cheng Q, Chen M, Chen X, Chen X, Jiang H, Wu H, et al. Novel long non-coding RNA expression profile of peripheral blood mononuclear cells reveals potential biomarkers and regulatory mechanisms in systemic lupus erythematosus. Front Cell Dev Biol. (2021) 9:639321. doi: 10.3389/fcell.2021.639321

PubMed Abstract | Crossref Full Text | Google Scholar

109. You Y, Zhao X, Wu Y, Mao J, Ge L, Guo J, et al. Integrated transcriptome profiling revealed that elevated long non-coding RNA-AC007278.2 expression repressed CCR7 transcription in systemic lupus erythematosus. Front Immunol. (2021) 12:615859. doi: 10.3389/fimmu.2021.615859

PubMed Abstract | Crossref Full Text | Google Scholar

110. Chen X, Luo X, Wei Y, Sun H, Dai L, Tangzhou Y, et al. LncRNA H19 induces immune dysregulation of BMMSCs, at least partly, by inhibiting IL-2 production. Mol Med. (2021) 27:61. doi: 10.1186/s10020-021-00326-y

PubMed Abstract | Crossref Full Text | Google Scholar

111. Zhang Y, Xie L, Lu W, Lv J, Li Y, Shao Y, et al. LncRNA MIAT enhances systemic lupus erythematosus by upregulating CFHR5 expression via miR-222 degradation. Cent Eur J Immunol. (2021) 46:17–26. doi: 10.5114/ceji.2021.105242

PubMed Abstract | Crossref Full Text | Google Scholar

112. Gao F, Tan Y, Luo H. MALAT1 is involved in type I IFNs-mediated systemic lupus erythematosus by up-regulating OAS2, OAS3, and OASL. Braz J Med Biol Res. (2020) 53:e9292. doi: 10.1590/1414-431x20209292

PubMed Abstract | Crossref Full Text | Google Scholar

113. Cao HY, Li D, Wang YP, Lu HX, Sun J, Li HB. Clinical significance of reduced expression of lncRNA TUG1 in the peripheral blood of systemic lupus erythematosus patients. Int J Rheum Dis. (2020) 23:428–34. doi: 10.1111/1756-185X.13786

PubMed Abstract | Crossref Full Text | Google Scholar

114. Liao Z, Ye Z, Xue Z, Wu L, Ouyang Y, Yao C, et al. Identification of renal long non-coding RNA RP11-2B6.2 as a positive regulator of type I interferon signaling pathway in lupus nephritis. Front Immunol. (2019) 10:975. doi: 10.3389/fimmu.2019.00975

PubMed Abstract | Crossref Full Text | Google Scholar

115. Lu C, Shao X, Zhou S, Pan C. LINC00176 facilitates CD4(+)T cell adhesion in systemic lupus erythematosus via the WNT5a signaling pathway by regulating WIF1. Mol Immunol. (2021) 134:202–9. doi: 10.1016/j.molimm.2021.02.018

PubMed Abstract | Crossref Full Text | Google Scholar

116. Liu X, Lin J, Wu H, Wang Y, Xie L, Wu J, et al. A novel long noncoding RNA lincRNA00892 activates CD4(+) T cells in systemic lupus erythematosus by regulating CD40L. Front Pharmacol. (2021) 12:733902. doi: 10.3389/fphar.2021.733902

PubMed Abstract | Crossref Full Text | Google Scholar

117. Xue Z, Cui C, Liao Z, Xia S, Zhang P, Qin J, et al. Identification of lncRNA linc00513 containing lupus-associated genetic variants as a novel regulator of interferon signaling pathway. Front Immunol. (2018) 9:2967. doi: 10.3389/fimmu.2018.02967

PubMed Abstract | Crossref Full Text | Google Scholar

118. Gao X, Liu L, Min X, Jia S, Zhao M. Non-coding RNAs in CD4(+) T cells: new insights into the pathogenesis of systemic lupus erythematosus. Front Immunol. (2020) 11:568. doi: 10.3389/fimmu.2020.00568

PubMed Abstract | Crossref Full Text | Google Scholar

119. Li LJ, Huang Q, Pan HF, Ye DQ. Circular RNAs and systemic lupus erythematosus. Exp Cell Res. (2016) 346:248–54. doi: 10.1016/j.yexcr.2016.07.021

PubMed Abstract | Crossref Full Text | Google Scholar

120. Yu HC, Tseng HH, Huang HB, Lu MC. Circ-CAMTA1 regulated by Ca(2+) influx inhibited pyruvate carboxylase activity and modulate T cell function in patients with systemic lupus erythematosus. Arthritis Res Ther. (2024) 26:185. doi: 10.1186/s13075-024-03422-6

PubMed Abstract | Crossref Full Text | Google Scholar

121. Mei HY, Liu J, Shen XP, Wu R. A novel circRNA, circRACGAP1, hampers the progression of systemic lupus erythematosus via miR-22-3p-mediated AKT signalling. Autoimmunity. (2022) 55:360–70. doi: 10.1080/08916934.2022.2073590

PubMed Abstract | Crossref Full Text | Google Scholar

122. Zhao X, Dong R, Zhang L, Guo J, Shi Y, Ge L, et al. N6-methyladenosine-dependent modification of circGARS acts as a new player that promotes SLE progression through the NF-κB/A20 axis. Arthritis Res Ther. (2022) 24:37. doi: 10.1186/s13075-022-02732-x

PubMed Abstract | Crossref Full Text | Google Scholar

123. He S, Du H, Wang Y, Shi X, Zhou Y. Hsa_circ_0010957 level is increased and sponges microRNA−125b in CD4(+) T cells of patients with systemic lupus erythematosus. Mol Med Rep. (2021) 23:469. doi: 10.3892/mmr.2021.12108

PubMed Abstract | Crossref Full Text | Google Scholar

124. Zheng F, Tan L, Zhang F, Li S, Lai Z, Xu H, et al. The circRNA-miRNA-mRNA regulatory network in plasma and peripheral blood mononuclear cells and the potential associations with the pathogenesis of systemic lupus erythematosus. Clin Rheumatol. (2023) 42:1885–96. doi: 10.1007/s10067-023-06560-5

PubMed Abstract | Crossref Full Text | Google Scholar

125. Zou H, Ma S, Li L, Xia X, Zhou Y, Zhang R. Downregulation of circular RNA ETS1 promotes SLE activity and inhibits Treg cell differentiation through miR-1205/FoxP3 molecular axis. Int Immunopharmacol. (2024) 128:111539. doi: 10.1016/j.intimp.2024.111539

PubMed Abstract | Crossref Full Text | Google Scholar

126. Yao R, Xu L, Cheng G, Wang Z, Liang R, Pei W, et al. Elevated expression of hsa_circ_0000479 in neutrophils correlates with features of systemic lupus erythematosus. Ann Med. (2024) 56:2309607. doi: 10.1080/07853890.2024.2309607

PubMed Abstract | Crossref Full Text | Google Scholar

127. Ouyang Q, Huang Q, Jiang Z, Zhao J, Shi GP, Yang M. Using plasma circRNA_002453 as a novel biomarker in the diagnosis of lupus nephritis. Mol Immunol. (2018) 101:531–8. doi: 10.1016/j.molimm.2018.07.029

PubMed Abstract | Crossref Full Text | Google Scholar

128. Luo Q, Ye Y, Zhang L, Gao Y, Rao J, Guo Y, et al. Hsa_circ_0044235 and hsa_circ_0001947 as novel biomarkers in plasma of patients with new-onset systemic lupus erythematosus. J Immunotoxicol. (2023) 20:2196453. doi: 10.1080/1547691X.2023.2196453

PubMed Abstract | Crossref Full Text | Google Scholar

129. Luo Q, Zhang L, Li X, Fu B, Guo Y, Huang Z, et al. Identification of circular RNAs hsa_circ_0044235 and hsa_circ_0068367 as novel biomarkers for systemic lupus erythematosus. Int J Mol Med. (2019) 44:1462–72. doi: 10.3892/ijmm.2019.4302

PubMed Abstract | Crossref Full Text | Google Scholar

130. Luo Q, Li X, Fu B, Zhang L, Fang L, Qing C, et al. Expression profile and diagnostic value of circRNAs in peripheral blood from patients with systemic lupus erythematosus. Mol Med Rep. (2021) 23:1. doi: 10.3892/mmr.2020.11639

PubMed Abstract | Crossref Full Text | Google Scholar

131. Luo Q, Zhang L, Xiong L, Fu B, Guo Y, Huang Z, et al. Peripheral blood circular RNA hsa_circ_0082688-hsa_circ_0008675 can be used as a candidate biomarker of systemic lupus erythematosus with renal involvement. Clin Exp Rheumatol. (2020) 38:822–33.

PubMed Abstract | Google Scholar

132. Li S, Zhang J, Tan X, Deng J, Li Y, Piao Y, et al. Microarray expression profile of circular RNAs and mRNAs in children with systemic lupus erythematosus. Clin Rheumatol. (2019) 38:1339–50. doi: 10.1007/s10067-018-4392-8

PubMed Abstract | Crossref Full Text | Google Scholar

133. Jiang Z, Li S, Jia Y, Wu Q, Chen X, Zhang M, et al. CircPTPN22 modulates T-cell activation by sponging miR-4689 to regulate S1PR1 expression in patients with systemic lupus erythematosus. Arthritis Res Ther. (2023) 25:206. doi: 10.1186/s13075-023-03150-3

PubMed Abstract | Crossref Full Text | Google Scholar

134. Miao Q, Zhong Z, Jiang Z, Lin Y, Ni B, Yang W, et al. RNA-seq of circular RNAs identified circPTPN22 as a potential new activity indicator in systemic lupus erythematosus. Lupus. (2019) 28:520–8. doi: 10.1177/0961203319830493

PubMed Abstract | Crossref Full Text | Google Scholar

135. Zhao X, Dong R, Tang Z, Wang J, Wang C, Song Z, et al. Circular RNA circLOC101928570 suppresses systemic lupus erythematosus progression by targeting the miR-150-5p/c-myb axis. J Transl Med. (2022) 20:547. doi: 10.1186/s12967-022-03748-2

PubMed Abstract | Crossref Full Text | Google Scholar

136. Li W, Fan R, Zhou C, Wei Y, Lin S, Wen S, et al. Circular RNA expression profile of systemic lupus erythematosus and its clinical significance as a potential novel biomarker. Genes Genomics. (2022) 44:1405–14. doi: 10.1007/s13258-022-01315-z

PubMed Abstract | Crossref Full Text | Google Scholar

137. Zheng F, Yu X, Tang D, Hong X, Zhang X, Liu D, et al. The identification of circular RNAs from peripheral blood mononuclear cells in systemic lupus erythematosus. BMC Med Genomics. (2021) 14:70. doi: 10.1186/s12920-021-00919-w

PubMed Abstract | Crossref Full Text | Google Scholar

138. Guo G, Wang H, Ye L, Shi X, Yan K, Lin K, et al. Hsa_circ_0000479 as a novel diagnostic biomarker of systemic lupus erythematosus. Front Immunol. (2019) 10:2281. doi: 10.3389/fimmu.2019.02281

PubMed Abstract | Crossref Full Text | Google Scholar

139. Luo Q, Zhang L, Fang L, Fu B, Guo Y, Huang Z, et al. Circular RNAs hsa_circ_0000479 in peripheral blood mononuclear cells as novel biomarkers for systemic lupus erythematosus. Autoimmunity. (2020) 53:167–76. doi: 10.1080/08916934.2020.1728529

PubMed Abstract | Crossref Full Text | Google Scholar

140. Zhang C, Wang X, Chen Y, Wu Z, Zhang C, Shi W. The down-regulation of hsa_circ_0012919, the sponge for miR-125a-3p, contributes to DNA methylation of CD11a and CD70 in CD4(+) T cells of systemic lupus erythematous. Clin Sci (Lond). (2018) 132:2285–98. doi: 10.1042/CS20180403

PubMed Abstract | Crossref Full Text | Google Scholar

141. Zhang C, Gao C, Di X, Cui S, Liang W, Sun W, et al. Hsa_circ_0123190 acts as a competitive endogenous RNA to regulate APLNR expression by sponging hsa-miR-483-3p in lupus nephritis. Arthritis Res Ther. (2021) 23:24. doi: 10.1186/s13075-020-02404-8

PubMed Abstract | Crossref Full Text | Google Scholar

142. Yang Y, Hsu PJ, Chen YS, Yang YG. Dynamic transcriptomic m(6)A decoration: writers, erasers, readers and functions in RNA metabolism. Cell Res. (2018) 28:616–24. doi: 10.1038/s41422-018-0040-8

PubMed Abstract | Crossref Full Text | Google Scholar

143. Shulman Z, Stern-Ginossar N. The RNA modification N(6)-methyladenosine as a novel regulator of the immune system. Nat Immunol. (2020) 21:501–12. doi: 10.1038/s41590-020-0650-4

PubMed Abstract | Crossref Full Text | Google Scholar

144. Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, et al. Anti-tumour immunity controlled through mRNA m(6)A methylation and YTHDF1 in dendritic cells. Nature. (2019) 566:270–4. doi: 10.1038/s41586-019-0916-x

PubMed Abstract | Crossref Full Text | Google Scholar

145. Deng LJ, Fang XY, Wu J, Li QR, Mao YM, Leng RX, et al. ALKBH5 expression could affect the function of T cells in systemic lupus erythematosus patients: A case-control study. Curr Pharm Des. (2022) 28:2270–8. doi: 10.2174/1381612828666220617154204

PubMed Abstract | Crossref Full Text | Google Scholar

146. Liu Y, Wang X, Huang M, Luo A, Liu S, Cai M, et al. METTL3 facilitates kidney injury through promoting IRF4-mediated plasma cell infiltration via an m6A-dependent manner in systemic lupus erythematosus. BMC Med. (2024) 22:511. doi: 10.1186/s12916-024-03735-y

PubMed Abstract | Crossref Full Text | Google Scholar

147. Tian Y, Tao K, Li S, Chen X, Wang R, Zhang M, et al. Identification of m6A-related biomarkers in systemic lupus erythematosus: A bioinformation-based analysis. J Inflammation Res. (2024) 17:507–26. doi: 10.2147/JIR.S439779

PubMed Abstract | Crossref Full Text | Google Scholar

148. Zhao H, Pan S, Duan J, Liu F, Li G, Liu D, et al. Integrative analysis of m(6)A regulator-mediated RNA methylation modification patterns and immune characteristics in lupus nephritis. Front Cell Dev Biol. (2021) 9:724837. doi: 10.3389/fcell.2021.724837

PubMed Abstract | Crossref Full Text | Google Scholar

149. Luo Q, Rao J, Zhang L, Fu B, Guo Y, Huang Z, et al. The study of METTL14, ALKBH5, and YTHDF2 in peripheral blood mononuclear cells from systemic lupus erythematosus. Mol Genet Genomic Med. (2020) 8:e1298. doi: 10.1002/mgg3.1298

PubMed Abstract | Crossref Full Text | Google Scholar

150. Zhao M, Zhou Y, Zhu B, Wan M, Jiang T, Tan Q, et al. IFI44L promoter methylation as a blood biomarker for systemic lupus erythematosus. Ann Rheum Dis. (2016) 75:1998–2006. doi: 10.1136/annrheumdis-2015-208410

PubMed Abstract | Crossref Full Text | Google Scholar

151. Zhang Bo, Liu L, Zhou T, Shi X, Wu H, Xiang Z, et al. A simple and highly efficient method of IFI44L methylation detection for the diagnosis of systemic lupus erythematosus. Clin Immunol. (2020) 221:108612. doi: 10.1016/j.clim.2020.108612

PubMed Abstract | Crossref Full Text | Google Scholar

152. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. The microRNA spectrum in 12 body fluids. Clin Chem. (2010) 56:1733–41. doi: 10.1373/clinchem.2010.147405

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: epigenetics, systemic lupus erythematosus, DNA methylation, histone modification, noncoding RNAs, RNA methylation, biomarkers

Citation: Zhou X, Zhou S and Li Y (2025) An updated review on abnormal epigenetic modifications in the pathogenesis of systemic lupus erythematosus. Front. Immunol. 15:1501783. doi: 10.3389/fimmu.2024.1501783

Received: 25 September 2024; Accepted: 09 December 2024;
Published: 06 January 2025.

Edited by:

Chris Wincup, King’s College Hospital NHS Foundation Trust, United Kingdom

Reviewed by:

Chunshu Yang, The First Affiliated Hospital of China Medical University, China
Jun Xue, Fudan University, China
Shangqing Ge, First Affiliated Hospital of Anhui Medical University, China

Copyright © 2025 Zhou, Zhou and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yaping Li, bHlwbGlzYUBjc3UuZWR1LmNu

†These authors share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.