Skip to main content
Therapeutic Advances in Chronic Disease logoLink to Therapeutic Advances in Chronic Disease
. 2023 Apr 3;14:20406223231153572. doi: 10.1177/20406223231153572

The emerging role of noncoding RNAs in systemic lupus erythematosus: new insights into the master regulators of disease pathogenesis

Mina Afrashteh Nour 1, Farid Ghorbaninezhad 2,3, Zahra Asadzadeh 4, Amir Baghbanzadeh 5, Hamidreza Hassanian 6, Patrizia Leone 7, Mahdi Jafarlou 8, Nazila Alizadeh 9, Vito Racanelli 10,, Behzad Baradaran 11,12,13,†,
PMCID: PMC10074641  PMID: 37035097

Abstract

Auto-immune diseases are a form of chronic disorders in which the immune system destroys the body’s cells due to a loss of tolerance to self-antigens. Systemic lupus erythematosus (SLE), identified by the production of autoantibodies in different body parts, is one of the most well-known examples of these diseases. Although the etiology of SLE is unclear, the disease’s progression may be affected by genetic and environmental factors. As studies in twins provide adequate evidence for genetic involvement in the SLE, other phenomena such as metallization, histone modifications, and alterations in the expression of noncoding RNAs (ncRNAs) also indicate the involvement of epigenetic factors in this disease. Among all the epigenetic alterations, ncRNAs appear to have the most crucial contribution to the pathogenesis of SLE. The ncRNAs’ length and size are divided into three main classes: micro RNAs, long noncoding RNAs (LncRNA), and circular RNAs (circRNAs). Accumulating evidence suggests that dysregulations in these ncRNAs contributed to the pathogenesis of SLE. Hence, clarifying the function of these groups of ncRNAs in the pathophysiology of SLE provides a deeper understanding of the disease. It also opens up new opportunities to develop targeted therapies for this disease.

Keywords: auto-immune disease, auto-immunity, circular RNAs, LncRNAs, micro RNAs, noncoding RNA, systemic lupus erythematosus

Introduction

Auto-immune diseases are a group of chronic disorders in which the immune system attacks the body’s own cells. Loss of immune tolerance to self-antigens causes this phenomenon. Although the cause of these diseases is not well known, they affect around 3% of the European and American populations. 1 Environmental and genetic variables, hormones, and infectious diseases have all been identified as major contributors to the development of auto-immune disorders in studies. 2 Some of the well-recognized auto-immune disorders include Addison’s disease, Hashimoto’s thyroiditis, multiple sclerosis (MS), Sjögren’s syndrome, systemic lupus erythematosus (SLE), and type I diabetes (T1D). 3 Although the symptoms of auto-immune diseases vary and depend on the type and location of the disease, the general indications of these disorders include fatigue, fever, lethargy, joint pain, and rash. Despite advances in studies related to auto-immune diseases, there is still no definitive treatment for this disorder. However, antimalarial drugs, steroidal and nonsteroidal anti-inflammatory drugs, immunosuppressive agents, and target therapies such as cytokine inhibitors [e.g. tumor necrosis factor-alpha (TNF-α) blockers] can all prevent the progression of auto-immunity.4,5 SLE is a histologically heterogeneous auto-immune disease defined by the development of autoantibodies directed against nuclear antigens. 5 This disease, which most frequently affects young to middle-aged women, has an incidence of 1 to 10 per 100,000 people annually. 5 Due to the uncertainty of SLE’s cause, several factors like genetic and environmental variables (epigenetic) have been studied in the incidence of SLE. 6 On the other hand, epigenetic factors, including methylation, histone modification, and especially ncRNAs, may be involved in the pathogenesis of SLE. ncRNAs are synthesized from a bigger portion of the genome that does not encode proteins and are implicated in gene expression and protein activity control. 7 MicroRNAs, long noncoding RNAs (LncRNA), and circular RNAs (circRNAs) are three of the most significant types of ncRNAs. MicroRNAs are small, endogenous, 19- to 25-nucleotide (nt) RNAs that inhibit the translation of specific mRNAs.811 Another type of ncRNA is LncRNAs which have more than 200 nt and do not convert to functional proteins. 12 CircRNAs are endogenous RNA molecules that can range in size from remarkably short (100 nt) to more than 4 kb and include exonic or intronic sequences of their parental genes. 13 According to studies, ncRNAs have a key role in B-cell activation and the development of SLE by the B-cell activation and the nuclear factor kappa light chain enhancer of activated B cells (NF-κB), type I interferons (IFN-I), and tumor growth factor (TGF) signaling pathways. 14 In this review, we will discuss the factors involved in the pathogenesis of SLE, especially microRNAs and LncRNAs, and their possible therapeutic use in the course of the disease.

Auto-immune disease

Auto-immune disorders are a group of long-lasting diseases that begin with the body’s inability to distinguish its cells from foreign cells, and the result is an attack and the development of an immune response against self-antigens. 1 Contrary to the low prevalence of auto-immune diseases (about 3%), their substantial influence on mortality cannot be ignored. 15 Normal immune responses often result in the removal of foreign antigens, e.g. pathogen-associated molecular patterns (PAMPs) and cancerous antigens from the body. Contrary to the physiological state, in auto-immune diseases, the immune system is unable to distinguish between self-antigens and foreign antigens. The result is the generation of excessive immune response to self-antigens and subsequent organ damage.3,16 Although a variety of factors may contribute to the generation of auto-immunity, immune tolerance breach is a crucial mechanism in the pathogenesis of this disease. 17 Central tolerance in the thymus and bone marrow is critical for immune system homeostasis. Before maturing and entering the bloodstream, developing lymphocytes in the thymus undergo positive selection in the cerebral cortex. It is noteworthy that in a healthy host, the thymic medulla negatively selects and eliminates lymphocytes with potential self-reactivity. 18 Peripheral tolerance or secondary selection of mature T cells occurs after exiting the thymus. In this phenomenon, most of the self-reactive T cells are eliminated or become anergic. 18 In addition, immature B cells are eliminated through a process named clonal deletion or clonal anergy if they express surface IgM that identifies common self-cell-surface antigens. 18 The ‘receptor editing’ approach permits autoreactive B cells to evade elimination. Peripheral tolerance can affect mature B cells as well. 18 The breakdown of these immune tolerance mechanisms and the presence of these potentially self-reactive T and B lymphocytes or their capacity to generate autoantibodies cause the onset of classical or pathological auto-immunity.2,19 However, even in healthy individuals, although peripheral and central tolerance is carefully maintained, some potentially autoreactive lymphocytes are present in the periphery, causing no symptoms of auto-immune disease.2,19 This state is named physiological auto-immunity, which is without evidence of clinical disease. The body’s natural antibodies, which aid in the removal of degraded self and foreign antigens to preserve homeostasis, are evidence of this condition. 20 Furthermore, despite the unknown factors influencing the immune response to this disease, the impact of environmental and genetic factors has been discussed in several studies.2,21,22 Numerous genetic polymorphisms, the majority of which are found in the regulatory regions of genes, have been implicated in the development of a variety of auto-immune diseases.23,24 For instance, among all the genes linked to auto-immune disorders, those with specific human leukocyte antigen (HLA) alleles had the highest and longest-standing correlations. 24 For instance, T1D (HLA-II: DQ2 and DQ8; HLA-I: HLA-A and DQB1*0602), rheumatoid arthritis (RA) (HLAII: DR4; HLA-III: TNF), auto-immune thyroid disease (HLA-II: DR3 and DR4), and psoriasis (HLA-I:Cw*0602, Cw1203, and HCP5) are highly correlated with certain HLA alleles. 25 Moreover, several auto-immune illnesses have been related to genetic variations in cytokines and cytokine receptors. Ankylosing spondylitis, Behcet’s disease, Crohn’s disease, psoriasis, and ulcerative colitis are all disorders where the IL23R gene has been found to have genetic variations. 26 In addition, rare cases of fulminant auto-immunity caused by genetic changes in a single gene have been reported. In light of this, auto-immune polyendocrine syndrome (APS) and immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) are instances of monogenetic auto-immune disorders. Auto-immune regulator (AIRE) and forkhead box P3 (FOXP3) mutations, respectively, cause these disorders directly.27,28 Growing evidence indicates that the development of auto-immune disease depends on environmental variables in addition to genetic predisposition. Nutrition, the microbiota, infectious processes, and xenobiotics such as cigarette smoke, pharmaceuticals, hormones, ultraviolet (UV) radiation, silica solvents, heavy metals, vaccinations, and collagen/silicone implants are among these environmental influences.2931 It has long been believed that infections can cause auto-immune disorders.32,33 For instance, MS has been linked to postmortem brain tissue showing indications of Epstein–Barr virus (EBV) infection, but not other inflammatory diseases. 34 Furthermore, it was demonstrated that patients with relapsing–remitting MS have been known to experience relapses due to systemic infections that increase myelin-specific T-cell responses. 32 RA and periodontal infections provide another evidence of the correlation between infections and auto-immune. 35 Moreover, the potential effect of the microbiome on local and systemic immune responses has also been reported. Immune responses to intestinal commensal microorganisms that are dysregulated and too aggressive lead to inflammatory bowel disease (IBD). 36 In addition, commensal microorganisms have been linked to auto-immune diseases including T1D. 36 UV radiation is a nonmicrobial environmental factor that has been linked to auto-immune disorders like cutaneous lupus. This association may be addressed by the concept that UV radiation causes the apoptotic death of many different cell types and enhances the burden of nuclear antigens, particularly if the dead cells cannot be effectively removed. 37 It has been proposed that groups of tolerance-promoting dendritic cells (DCs) preserve peripheral tolerance to tissue antigens via low-level spontaneous cell death in tissues. This system may become easily overwhelmed in lupus patients because of a genetic predisposition that renders them unable to maintain tolerance in the presence of constant UV exposure. 38 Several causes can contribute to the development of auto-immune diseases. For instance, it was noted that there is presumably an imbalance between effector T cells and functional regulatory T cells (Tregs) in T-cell-dependent inflammatory auto-immune disorders. 2 This pathogenic imbalance was supported by mouse models of auto-immunity. 39 Moreover, human auto-immune diseases may also be initiated by decreased numbers of functional Tregs, or by the resistance of effector T cells to regulation. 2 Antigen-presenting cells (APCs) can present denatured or extracellular peptides atypically, resulting in peptide/major histocompatibility complexes (MHCs), that are distinct from those typically generated inside APCs, and are therefore capable of activating T cells that may be potentially pathogenic. 40 The identification of conformational isomers of the peptide/MHC or a difference in the binding register of a peptide within the groove of the MHC molecule may be the cause of this unusual activation of potentially self-reactive T cells.41,42 Besides altered peptide/MHC recognition, auto-immunity can be triggered by an early innate immune response. 43 In light of this, the lack of ubiquitin-modifying enzyme A20 leads to lethal auto-immunity resulting from unregulated Toll-like receptor (TLR) signals mediated by MyD88. 44 Also, due to enhanced B-cell identification of nucleolar antigens in Yaa mice with a genetic duplication of TLRs-7, these mice exhibit a spontaneous SLE-like syndrome. 45 SLE, one of the most prevalent auto-immune diseases, is a multisystem auto-immune disorder that targets the body’s tissues and results in significant organ damage and inflammation. Despite much investigation, the etiology of SLE is still unclear. Both genetic and environmental variables have been related to the pathogenesis of SLE. In the following sections, we will discuss this disease in detail.

SLE

SLE is one of the most prevalent auto-immune diseases that can affect any organ and part of the body and cause irreversible complications. The literal meaning of lupus in ancient Latin goes back to the wolf, and the name of this disease is because in the 13th century, for the first time, the physician Rogerius likened the facial lesions of a person with this disease to imitating the bite of a wolf. 46 Since then, many researchers have worked on it and have used many descriptions of its skin symptoms, including erythema centrifugum or lupus discoid and disease with butterfly lesions on the face.4648 Other pathological symptoms and features of this chronic disease were later identified, including central nervous system (CNS) involvement, inflammation, and vascular abnormalities such as vasculitis and immune complex deposition.49,50 Essentially, the clinical manifestations of lupus imitate a viral syndrome with signs of weight loss, fatigue, and low-grade fever with other skin, kidney, respiratory, cardiovascular, CNS, and gastrointestinal indications. 50 Epidemiologically, the disease is more common in adult women during the reproductive period, about 10:1 compared with men. Despite the lack of a comprehensive global method for identifying the true prevalence and incidence of the disease, the yearly incidence of SLE is estimated at 1–10 per 100,000, and the prevalence is 20–70 per 100,000 per year.5154

SLE has a complex etiology that involves environmental factors and genetic predisposition (Figure 1). As with other auto-immune diseases, loss of immune tolerance to self-antigens along with sustained autoantibody generation are basic pathologic processes in the development of SLE. 50 The tolerance mechanisms in SLE patients have been found to exhibit several obstacles, including defects in the regulation of mature B cells in peripheral tissues, receptor editing processes, and deletion of immature B cells in the bone marrow. 55 Also, a failure in the early B-cell tolerance checkpoints may be a factor in the establishment of SLE since mature naïve B cells in patients with this condition can generate autoantibodies even before the encounters with antigens. 55 Besides, another fundamental concept in SLE pathogenesis is an imbalance between apoptotic cell generation and apoptotic material removal. During apoptosis, the cell membrane creates blebs that pinch off from the cell and include fragmented cellular material, particularly nuclear antigens, which are normally inaccessible to the immune system. 56 Daily, neutrophils in humans undergo apoptosis; exposure to UV radiation, infections, and toxins can increase the apoptosis of these cells, which are all established to be linked to SLE. 57 Furthermore, studies of peripheral blood samples from lupus patients reveal pyroptosis, a form of cell death, as a new pathway for the disease. Accordingly, dying and dead cells can produce a large amount of immune complex components in the blood. Consequently, pyroptosis can significantly affect the auto-immune response in these diseases. Gasdermin D (GSDMD) has recently been identified as the ultimate pyroptosis tormentor in the activation of inflammatory diseases and hence can play an important role in the pathogenesis of SLE (NCT03984227). All of these apoptotic fragments are often quickly removed and are inaccessible to the immune system. However, abnormalities in the removal of apoptotic cells can lead to the development of SLE by immune dysregulation. In this regard, these auto-antigens are recognized as nonself by the innate immune system cells in SLE due to increased cell death and reduced removal of cellular debris. 58 For instance, nucleic acid recognition receptors, like TLRs, can be triggered by persistent apoptotic debris containing nucleic acids in order to initiate an inflammatory response. 59 In addition to the identification of viral infections and protection against intracellular bacteria, nucleic acid recognition receptors are linked to the generation of IFN-I (IFNα/β). 57 The pathogenesis of SLE is currently thought to be highly influenced by abnormalities in these pathways, which both increase disease susceptibility and directly cause monogenic types of SLE. 57 The differentiation of B cells and loss of tolerance are stimulated by IFN-I. 57 Nucleic acid recognition receptor–ligand interactions not only cause the production of IFN-I but also promote the generation of additional innate immune mediators like IL-1, IL-6, and TNF-α. These mediators stimulate APCs, resulting in enhanced antigen presentation to T cells. 60 Both surface IgM receptors for proteins complexed with nucleic acids and direct antigen detection by B lymphocytes allow them to respond to nucleic acids. 57 Also, in SLE patients, due to the defects in the removal of auto-antigens, nuclear material, and modified auto-antigens accumulate on germinal center follicular dendritic cells (FDCs), and autoreactive B cells are exposed to these modified auto-antigens by these FDCs. 61 After being triggered, these B cells mature, proliferate, and start secreting more antibodies, which strengthens the immune system’s adaptive response. 57 Besides, TNF-α, IL-4, IL-6, and IL-10 promote auto-antibody secretion and inflammation. 57 As most of the auto-antibodies found in SLE are high-affinity, somatically mutated IgG, this implies that the germinal centers, where T cells aid in class switching, are the likely site of their origination.49,62 T cells are therefore crucial to the development of the pathogenesis of SLE. In this regard, antigen uptake by APCs is degraded and presented to T cells as peptides–MHC complexes to initiate their responses. The stimulation and differentiation of T lymphocytes are also influenced by the presence of soluble mediators. 58 Pathogenesis of SLE has been linked to changes in the number of cytokines secreted by T cells, including elevated cytokine secretion of type 1 T helper (Th1) (IFN-γ, IL-2), type 2 T helper (Th2) (IL-5, IL-13), and type 17 T helper (Th17) (IL-6, IL-17A, IL-21) and decreased Treg responses, including diminished TGF-β and IL-10 production.6365 Increased T-follicular helper (Tfh) cell responses promote antibody-producing B cells with a high affinity for clonal expansion in germinal centers. 66 For example, in SLE patients, the production of pathogenic IgG antibodies with a high affinity for double-stranded DNA (anti-dsDNA) results from the interaction of B and T lymphocytes.49,62 Taken together, lymphocyte signaling, IFNα/β generation pathways, nucleic acid sensing, inefficient removal of biological debris like apoptotic cells, and neutrophil extracellular traps are all key contributors to loss of self-tolerance and tissue damage in SLE patients. Genome-wide association studies (GWAS) also support the involvement of this pathway in the pathogenesis of SLE.67,68 As stated, environmental factors affect the pathogenesis of SLE. In this regard, SLE has long been acknowledged to be influenced by hormones and UV radiation.69,70 It has been estimated that 90% of SLE cohorts consist of women, and estrogen and prolactin are known to enhance immune responses through a variety of mechanisms.71,72 It is believed that UV light stimulates the immune system by promoting apoptosis. 73 The role of EBV and cytomegalovirus infections has also been shown in the pathogenesis of SLE. 74 Moreover, there are many investigations on the correlation between SLE and genetics, and the higher incidence in monozygotic twins (24–58%) than dizygotic twins (2–5%) is proof of the accuracy of this hypothesis. 6 Also, SLE is highly correlated with certain HLA alleles (HLA-II: DR3, DR2, and DR8; HLA-III: SCIVaL, CFB, RDBP, DOM3Z, STK19C4A, and C4B). 25 In addition, this disease is influenced by epigenetic mechanisms such as noncoding RNAs (ncRNAs) and DNA methylation (the following sections will provide more details). 62 Due to the complexity of SLE and its treatment methods, these patients must have a comprehensive care plan. These treatments vary depending on the clinical manifestations and may include antimalarial drugs, glucocorticoids, immunosuppressants, and biological drugs. Also, nonsteroidal anti-inflammatory drugs (NSAIDs) can be used to limit inflammation or ache. 75

Figure 1.

Figure 1.

Factors that contribute to SLE pathogenesis. SLE pathogenesis is influenced by a variety of variables, including genetics, environment, hormones, and epigenetics. Each of these factors, in turn, triggers its own antibodies against the immune system. By presenting these antibodies on lymphocytes and producing various immunoglobulins, inflammatory pathways are activated and destroy various tissues, including the kidneys, skin, lungs, brain, and heart.

C, complement; CD, cluster of differentiation; HLA, human leukocyte antigen; Ig, immunoglobulin; IL, interleukins; MHC, major histocompatibility complex; ncRNAs, noncoding RNAs; self-dsDNA, self-double-stranded DNA; SLE, systemic lupus erythematosus; TCR, T-cell receptor; TNF-α, tumor necrosis factor-alpha.

Different mechanisms involved in the development of SLE

Lupus is an auto-immune and heterogeneous disease that has been labeled a mysterious and cruel disease in the United States due to its complexity and ambiguity. 76 Although the definite cause of this complex multifactorial disease has not yet been identified, several variables such as genetic, epigenetic, ethnic, immune, hormonal, and environmental factors are involved in its progression. As a result, each of these factors causes abnormal activation of T lymphocytes and other immune cells, and eventually, the accumulation of these cells causes damage to the target organs.77,78 The higher incidence of SLE in first-degree relatives (5.87%) and high heritability (43.9%) is also a reason for the genetic influence on this disease. According to the available evidence and research, a defect in one gene alone cannot cause disease, and the cooperation of several genes and alleles is required for its occurrence. However, it has been shown that defects in complement component genes such as 1q (C1q), subcomponent A (C1QA), C1QB, C1QC, and DNA repair genes like exonuclease 1 (TREX1), or deoxyribonuclease 1-like 3 (DNASE1 L3) can also trigger the monogenic forms of the lupus-like disease.78,79 According to relevant studies in GWAS, HLA genes are found on chromosome 6 and in MHC regions, which are high polymorphic areas. Because of the diversity in its genes, especially MHC II, they can have a greater odds ratio (OR) of SLE involvement. The presentation of specific peptides to CD4+ T cells to remove self-reactive lymphocytes (negative selection) and stimulate the production of T regs by MHC II occurs during the maturation of T cells in the thymus. As a result, if mutations occur in these areas, this process will be disturbed.8084 Considering the effect of gender on the SLE, it can be noted that because of the effect of estrogen on humoral responses and maintaining the self-response of B cells, the ratio of women to men is 10:1. Compared with estrogen, testosterone appears to suppress anti-DNA antibodies and somehow prevent the development of SLE.8587 Furthermore, environmental factors encompass a wide range of elements that influence the progression of SLE. Epstein–Barr virus, light exposure, and smoking are examples of biological, physical, and chemical stimuli that increase the development of anti-dsDNA antibodies and cell necrosis, respectively. In addition, certain medications can also cause iatrogenic lupus. 76 Although these factors are significant in the development of SLE, hereditary background and regulation of susceptibility genes are also required. Environmental factors affect the growth of self-reactive lymphocytes and interfere with the processing and presentation of antigens. They also expose B and T lymphocytes to microbial agents and trigger the polyclonal activation of these cells. With the cooperation of these factors, the incidence of SLE and its consequent damages will increase. 87 Epigenetic changes also have a significant influence on genetic diversity in many diseases, including SLE. Alterations such as DNA methylation, histone modifications, and ncRNAs are all included and determine how regulatory disturbance happens in SLE. 77 Global DNA hypomethylation is present in human T lymphocytes of active SLE patients, particularly in those with lupus nephritis.88,89 Since DNA methylation is normally restrictive, the result of this hypomethylation is frequently the upregulation of genes. For example, promoter hypomethylation in this disorder causes the overexpression of genes like CD11a and CD70 in T cells. 88 Furthermore, IFN-stimulated genes (ISGs) were notably hypomethylated when evaluated on a genome-wide basis in individuals with SLE. 89 The extracellular signal–regulated kinase (ERK)1/2 is essential for the induction of T-cell responses. Both independent and dependent responses to the T-cell receptor (TCR) trigger the ERK pathway in T cells. 90 ERK signaling pathway also regulates the expression of T-cell DNA methyltransferase (Dnmt). 90 As the ERK signaling pathway regulates the expression of T-cell Dnmt, 90 defects in this signaling pathway can lead to epigenetic abnormalities in T cells, such as a decrease in DNA methylation at the genome level. 90 In line with this, it has been shown that defective T-cell ERK signaling is associated with the pathogenesis of SLE in the mouse model of the disease. 88 The generation of anti-dsDNA antibodies is induced by defective ERK signaling in T cells. 88 Similar to T cells in human lupus, reduced ERK pathway activation in T cells causes upregulation of the methylation-sensitive genes CD11a and CD70 and reduced expression of Dnmt-1. 88 The degree of T-cell ERK signaling impairment as well as the anti-dsDNA antibody titer is positively correlated with disease severity in lupus patients.91,92 Furthermore, autoreactivity and altered cytokine release may both be caused, directly or indirectly, by aberrant lupus T-cell DNA methylation.92,93 Another crucial signaling pathway in the immunopathogenesis of lupus is phosphoinositide 3-kinase (PI3 K)/Akt (Ak strain transforming)/mammalian target of rapamycin (mTOR) pathway. Cellular proliferation and expression of inflammatory cytokines are greatly influenced by this signaling pathway. 94 SLE patients’ CD4+ T cells had higher Akt expression and phosphorylation levels than healthy donors’ CD4+ T cells, which suggests that PI3 K and mTOR activity have been elevated. 95 There has also been evidence of increased p70S6k (one of the mTOR substrates) activity in SLE CD4+ T cells. 95 Also, when compared with healthy donors’ peripheral blood mononuclear cells (PBMCs), SLE patients’ PBMCs had significantly greater levels of Akt phosphorylation and lower levels of p27kip1 expression. 96 Increased Akt activity and decreased p27kip1 expression appear to be factored in the passage of SLE lymphocytes through the G1/S cell cycle checkpoint. As a result, SLE lymphocytes gather in the G2/M and S-cell-cycle stages as they prepare to undergo proliferation. 96 The association between SLE pathogenesis and ncRNAs is now well established. Contrary to previous notions of these RNAs as evolutionarily junk, they have a substantial role in the molecular processes and pathogenesis of several disorders by forming a large part of the whole transcriptome.33,34 One of the most important ncRNAs associated with the occurrence of SLE and its pathogenesis is microRNAs. These small 22-nt ncRNAs are responsible for the negative regulation of gene expression that binds to target mRNA. In other words, it degrades or inhibits RNA translation and silences it after attaching to the target.76,97 So, ncRNAs affect pivotal signaling pathways involved in SLE pathogenesis, such as NF-κB, IFN-І, and TGF pathways. 14 In addition, the different roles of microRNAs in signal transducer and activator of transcription (STAT) pathway regulation are central. Due to the incorrect regulation of this pathway in SLE patients and its importance in Th17 differentiation, an increase in IL-17 and Th17 rates is observed. 98 Furthermore, ncRNA influenced the activation and differentiation of CD4+ T cells. Thus, dysregulation of ncRNAs such as miR-21, miR-155, miR-31, miR-29a, miR-126, miR-142-3p/5p, and miR-183 C, as well as LncRNAs in CD4+ T cells or PBMCs, has been observed to increase the incidence of SLE through direct and indirect regulation of CD4+ T cells. This is followed by a high proliferation of Th1/Th17/ Tfh cells, which reduces the number of Treg cells and, in this way, contributes to the development of SLE. 99 Also, in a clinical study of pregnant women with SLE, it was found that there was a 40% increased risk of flare-up symptoms with the onset of pregnancy, even in stable and asymptomatic patients. Epigenetic modification and changes in the expression of microRNAs during pregnancy are supposed to be the cause of increased signs and symptoms as well as the risk of flares in SLE (NCT02350491). Besides, recent studies have attempted to elucidate the involvement of circRNAs in SLE via microarrays and high-throughput sequencing. Their results have revealed the involvement of these molecules in the pathogenesis of SLE.100,101 The ncRNAs help identify and differentiate SLE patients besides performing well in diagnosing disease complications. Despite a thorough investigation of coding regions and their association with single-nucleotide polymorphism (SNP) in the pathogenesis of SLE, studies related to noncoding regions as substantial risk factors for SLE need further investigations to be more clarified.14,102Figure 1 illustrates the pathogenesis-related factors for SLE. In the following sections, we will quite describe ncRNAs. However, there is a need for more studies to clearly understand the importance of these RNAs in SLE’s pathogenesis.

The role of microRNAs in the development of SLE

MicroRNAs are short, endogenous, and ncRNAs 19- to 25-nt long, which perform various functions including cell cycle regulation, differentiation, proliferation, apoptosis, stress tolerance, energy metabolism, and immune response.811 Targeting specific mRNAs and destroying or suppressing their translation is another regulatory role of these ncRNAs.9,11 The first short RNA, known as lin-4, was identified in 1993 while screening nematodes. Coincidentally, in the same year, the regulation of lin-14 was also determined by lin-4 and showed the regulatory function of these RNAs for the first time.103,104 To this point, 1917 pre-microRNAs and 2654 mature microRNAs have been identified in Homo sapiens, whose target sites have been predicted by 60% of human coding genes. Mature microRNA is transcribed from the primary microRNA (pri-microRNA) during several stages of its biogenesis. They often originate from introns or LncRNAs and are copied by RNA polymerase II. 105 Numerous studies have shown an association between microRNA expression in peripheral blood cells, body fluids, and damaged tissues of SLE patients with severe damage to various organs in these individuals. For instance, in a study on 100 patients with SLE aged 18 and above who were cured with cyclophosphamide, hydroxychloroquine, and hydrocortisone, the expression levels of microRNAs, e.g. miR181a, miR196a, and miR21, were evaluated for 3 months to record changes in their expression (NCT02756546). According to these findings, microRNAs are significant biomarkers in diagnosing auto-immune diseases like SLE. Improper regulation of microRNAs in T cells can cause immunodeficiency and ultimately lead to auto-immune disease. 106 As a result, investigating the association between microRNAs and auto-immune diseases has a lot of importance nowadays. Table 1 provides a list of microRNAs related to SLE disease. Besides, the role of microRNAs in the pathogenesis of SLE is depicted in Figure 2.

Table 1.

MicroRNAs related to SLE disease.

miRNA Number of clinical samples Biological sample Target gene Result References
MiR-181 24 SLE patients and 28 healthy people T cells IL2, IL4, IL6, IL10, IFN-ɤ, (UC-MSC) can upregulate miR-181a, which may help improve SLE. 107
100 SLE patients and 40 healthy individuals Plasma ESR, CRP, C3, C4 MiR-181a levels in plasma are significantly higher in SLE patients. 108
402 SLE patients and 430 age- and gender-matched healthy controls Plasma IFNA1 Downregulation of miR-181b was revealed in SLE patients due to rs322931 CT/TT genotype compared with normal individuals. 109
80 patients with SLE and 104 control group serum MTMR3 The association between miR-181a and MTMR3 enhances autophagy in SLE patients. 110
202 patients with SLE and 299 control T lymphocytes (rs16927589 T>C, rs77418916 A>T, and rs8108402 C>T Higher association of the rs8108402 C>T polymorphism on the miR-181c/d gene promoter compared with other genotypes in SLE was reported. 111
70 SLE patients and 30 control groups Plasma IL-10, C3, C4 MiR-181a expression was increased in severe patients compared with mild patients. 112
MiR-21 70 SLE patients and 30 control groups Plasma IL-10, C3, C4 The expression of miR-21 was considerably greater in severe patients compared with mild individuals. 112
50 SLE, 30 healthy patients PBMC HIF-1a Patients with active SLE had higher levels of miR-21p expression than the control group. 113
45 SLE patients, 30 control PBMC CD4+ T cells MiR21 levels were significantly higher in CD4+ T cells from SLE patients with low complement component 3 (C3) levels compared with those with normal complement C3 levels. 114
MiR-196a 70 SLE patients and 30 control groups Plasma IL-10, c3, c4 When compared with the healthy group, miR-196a demonstrated the greatest upregulation in SLE patients. 112
MiR-98 48 patients with SLE, 39 healthy controls PBMC Fas, caspases, CD4+ T cells Reduced miR-98 expression causes death in CD4+ T SLE cells by altering the Fas-mediated apoptosis signaling pathway. 115
41 SLE patients and 20 healthy controls PBMC IL-6, TNF-α, IL-8, IL-1β, and IL-10 MiR-98 was discovered to be downregulated in SLE patients’ PBMCs, which is inversely related to IL-6 levels. 6
MiR-155 40 SLE patients, 13 healthy controls Urine TNF-α MiR-155 was shown to be downregulated in serum and urine samples from SLE patients compared with controls. 116
40 SLE patients, 32 healthy controls PB MiR-155 overexpressed in PB of SLE patients compared with the control group. 117
Mouse (12 injected NPA, 4 control) serum IFN1/2 iR-155-5p displayed downregulated expression in all murine-like models. 118
60 SLE patients and 40 healthy controls Serum/urine CRP, ANA, anti-dsDNA, C3 and C4, liver function tests, and renal function tests High expression of miR-155 in SLE can be used as a biomarker in disease progression. 119
40 patients with SLE and 30 healthy controls Serum/urine Complete blood count, serum complement levels, serum creatinine, urea, proteinuria, serum high-sensitivity C-reactive protein (hsCRP), and SLEDAI The SLE group had lower levels of miR-155 in their blood than the control group. 120
24 SLE, 75 control group Serum TLRs When compared with the control group, SLE patients have considerably higher levels of miR-151 expression. 121
MiR-210 50 SLE, 30 healthy patients PBMC HIF-1a In clinical practice, the miR network in SLE can be employed as a diagnostic or prognostic indicator. 113
35 patients with SLE, 35 controls PBMC HIF-1a In individuals with SLE, miR-210 expression levels were significantly higher than in healthy controls. 122
MiR-126 88 SLE patients, 30 control plasma Alpha interferon (aIFN) In SLE, plasma levels of miR-126 were considerably greater than in controls. 123
MiR-146a 88 SLE patients, 30 control Plasma Alpha interferon (aIFN) In comparison with the control group, SLE patients had lower levels of miR-146a. 123
40 patients with SLE and 30 healthy controls Serum/urine Complete blood count, serum complement levels, serum creatinine, urea, proteinuria, serum high-sensitivity C-reactive protein (hsCRP), and SLEDAI MiR-146a levels in the serum and urine reduced and rose in the SLE group, respectively, as compared with the control group. 120
39 healthy donors and 101 SLE patients PBMC I-IFN, CCL2, and CXCL10 STAT1 and miR-146a upregulation appears to play a significant role in anemia in SLE patients. 124
10 healthy female donors and 10 female SLE patients Serum MSC senescence MiR-146a expression was shown to be lower in SLE patients’ serum exosomes compared with healthy controls. 125
MiR-146a-5p 42 female SLE patients and 39 matched healthy individuals Serum C4 MiR-146 expression was shown to be increased in individuals with SLE symptoms such as retinopathy. 126
MiR-200a-3p Mouse (12 injected NPA, 4 control) serum IFN1/2 MiR-200a-3p was shown to be increased in all murine-like models. 127
MiR-125b 12 SLE patients and 12 control group PBMC ATG7, Beclin-1, and LC3 II MiR-125b expression was decreased in the PBMCs of SLE patients compared with healthy controls. 128
MiR-29b 42 patients and 35 healthy controls PBMC CircIBTK, C3 MiR-29b expression was increased in SLE patients. 129
MiR-29a 106 patients with SLE, 43 healthy group PBMC CRKL MiR-29a expression was shown to be reduced in SLE patients. 130
MiR-199-3p 29 patients with SLE and 25 healthy subjects PBMC Poly (ADP-ribose) polymerase-1 miR-199-3p was found to be overexpressed in SLE patients. 131
MiR-125a-3p 10 female MRL/MPJ-Fas lpr/J mice and 10 BALB/C female mice DMEM media, added with FBS (10%) IL-17 The levels of miR-125a-3p in the kidneys of MRL/MPJFas lpr/J mice were lower than in BALB/C mice. 132

ANA, anti-nuclear antibodies; anti-dsDNA, anti-double-stranded DNA; BALB/C, Bagg Albino; C, complement; C-C motif, the chemokine; CCL2, ligand 2; circRNAs, circular RNAs; CRKL, Crk-like protein; CRP, C-reactive protein; CXCL10, C-X-C motif chemokine ligand 10; DMEM, Dulbecco’s Modified Eagle Medium; ESR, erythrocyte sedimentation rate; FBS, fetal bovine serum; HIF-1, hypoxia-inducible factor; IFNA1, interferon-alpha receptor; IL, interleukins; LC3, autophagy-related 7, microtubule-associated protein 1A/1B-light chain 3; MRL/MPJ-Fas lpr/J, Murphy Roths large/homozygous for the lymphoproliferation spontaneous mutation; MSCs, mesenchymal stem cells; MTMRT3, myotubularin-related protein 3; NPA, nonbilayer phospholipid arrangements; PBMC, peripheral blood mononuclear cell; SLE, systemic lupus erythematosus; SLEDAI, Systemic Lupus Erythematosus Disease Activity Index; TLRs, Toll-like receptors; TNF-α, tumor necrosis factor-alpha; UC-MSC, umbilical cord mesenchymal stem cells.

Figure 2.

Figure 2.

Role of microRNAs in the SLE. Many microRNAs, including miR-181, miR-146, miR-155, etc., are involved in the pathogenesis of SLE. High or low expression of any of these ncRNAs regulates multiple signaling pathways and is somehow involved in the pathogenesis of this auto-immune disorder.

C, complement; HIF-1, hypoxia-inducible factor; IFNA1, interferon-alpha receptor; MiR, microRNA; MRL/MPJ-Fas lpr/J, Murphy Roths large/homozygous for the lymphoproliferation spontaneous mutation; NF-κB, nuclear factor-kappa B; PBMC, peripheral blood mononuclear cell; STAT, signal transducer and activator of transcription; TLRs, Toll-like receptors; TNF-α, tumor necrosis factor-alpha; UC-MSC, umbilical cord mesenchymal stem cells.

MiR-181

MiR-181 is a protected family consisting of miR-181a, miR-181b, miR-181c, and miR-181d, in which expression of miR-181a and miR-181b is generally observed in the brain, bone marrow, spleen, and thymus. These microRNAs are located on chromosomes 1, 9, and 19 in clusters. They have a main part in the regulation of B and T cells in auto-immune diseases. 133 MiR-181a, being expressed in several tissues, inhibits the proliferation of macrophages and induces apoptosis in them. Besides, by inhibiting the differentiation of Th1 cells, miR-181a and miR-181b differentiate regulatory T lymphocytes, which are responsible for hindering inflammation. 108 As miR-181a has a central function in T lymphocyte maturation and development, its aberrant expression may have an association with auto-immune disorders such as SLE. 108 A study was conducted by Lee et al. to evaluate miR-181a levels in active SLE, inactive SLE, and healthy groups. They discovered that the SLE group had greater serum levels of miR-181a than the normal group. Also, the levels of miR-181a in inactive SLE patients were upper than in both other groups. 108 On the other hand, overexpression of miR-181a in the peripheral blood of SLE patients has also been observed. 134 Yong-Ling and colleagues showed that miR-181b facilitates SLE development by directly targeting molecular IFN alpha 1 (IFNA1). For this purpose, they also investigated two SNPs (rs1332190 and rs10811543) located in the IFNA1 promoter region to analyze gene interactions. Interestingly, they found an association between miR-181b rs322931 C and SLE and its uptrend. Although IFNA1 rs1332190 and rs10811543 alone were not significantly related to SLE, a combined analysis of these three factors simultaneously augmented the risk of disease progression. Therefore, He et al., 109 finding a correlation between miR-181b expression and rs322931 and mentioning a downregulation in miR-181b levels in SLE patients due to rs322931 CT /TT genotype compared with normal individuals once again noted the importance of ncRNAs in SLE. Due to the effects of SNPs and their correlation with microRNAs, the association of autophagy-related gene, MTMR3 SNP rs12537, which is the main target gene of miR-181-a, has been investigated with SLE progression. Based on the results reported by Senousy et al., MTMR3 rs12537 TT is considered a risk factor in SLE with a higher risk of autophagy and has an unfavorable prognosis with it. Notably, miR-181a plays a crucial role in the development of SLE. 110 Remarkably, a case–control study also investigated three miR-181 cluster polymorphisms and their association with SLE in the Chinese population. According to this study, Wang et al. reported a higher association of the rs8108402C>T polymorphism on the miR-181c/d gene promoter compared with other genotypes in SLE. 135 Contrary to these findings, in another study, it was found that miR-181a downregulation occurs in T lymphocytes of SLE patients and indicated that using umbilical cord mesenchymal stem cells (UC-MSCs) can upregulate miR-181a, which may help improve the condition of SLE patients. 107

MiR-98

MiR-98, with a size of 119 bases, originates from the let-7 family and is involved in Niemann–Pick disease, type C1, and head and neck squamous cell carcinoma. Moreover, the role of let7/miR-98 in cell proliferation, differentiation, apoptosis, and angiogenesis has been discussed in several studies.136141 We can highlight the influence of mir-98 in SLE because of the increased apoptosis of T cells in SLE and the association of mir-98 with this phenomenon. It is noteworthy that the amount of mir-98 in SLE patients’ CD4+ T lymphocytes was lower than in normal people. 115 Therefore, this deficiency can be attributed to a defect in the regulation of the apoptosis pathway in SLE patients. As a result, mir-98 can be utilized to improve the condition of these patients by modulating apoptotic pathways while preserving lymphocyte homeostasis. 115 Several studies found a negative correlation between mir-98 and IL-6.142,143 By binding to 3′UTR of IL-6 mRNA, mir-98 negatively regulates this cytokine. The result is increased PBMC proliferation and suppression of the inflammatory factors production such as TNF-α, IL-8, IL-1β, and IL-10 in PBMC. Also, due to the vital role of STAT3 in hindering TNF-α, the ability of STAT inhibitor, S31-201, to revoke the function of both mir-98 inhibitors and high IL-6 expression was realized. According to these findings, mir-98 could be a key target in the treatment of SLE and other IL-6-related diseases. 6

MiR-155

MiR-155, encoded by MIRHG155, or originally the B-cell integration cluster (BIC) gene, matures in the third exon of this cluster, located in the nonencoding part of chromosome 21.144,145 Due to the pivotal role of this ncRNA in regulating crucial signaling pathways, its inappropriate expression can lead to a variety of cancers and auto-immune disorders such as RA, MS, and SLE. 146 The serum cell–free miR-155 expression profile was considered in another study in mixed connective tissue disease (MCTD), SLE, scleroderma and systemic sclerosis (SSc), and RA disease. Its significant expression in SLE in comparison with other disorders can indicate the substantial role of this miR in SLE. 121 MiR-155 has also been documented to have a higher expression in the peripheral blood of SLE patients than normal individuals, which may show renal injuries in these patients. 119 Another study by Shumnalieva et al. investigated the correlation between miR-155 and its changes in whole blood. According to this study, the expression of miR-155 in SLE patients compared with the control group was reported to be 50%, indicating that more studies are needed in this regard. 117 MiR-155 deficiency has also been linked to lesions and complications reduction in Fas LPR/LPR mice with SLE. 147 In contrast, Wang et al. 120 reported a decrease in the expression of miR-155 in serum and urine samples of patients with SLE compared with controls, which was consistent with the level of kidney function of these people. Due to the positive correlation between serum miR-155 levels and lymphocyte count, they have proposed that lymphocytes are the major source of serum miR-155 in SLE patients. Hence, miR-155 expression was likely reduced as a result of the decrease in the serum’s lymphocyte count. In addition, a nonsignificant decrease in the expression of miR-155 in SLE patients compared with the control group was also reported in the study of Latini et al. 148 They have suggested that this may be associated with miR-155’s regulatory roles in immune cells such as T and B cells.

MiR-146

MiR-146a is another type of ncRNAs located on human chromosome 5 in the second exon of the LOC285628 gene. The association of miR-146a with the pathogenesis of several diseases, including auto-immune disorders such as SLE, RA, and Sjögren’s syndrome, has been reported in various studies. 149 In this line, a research was conducted on 42 female SLE patients and 39 matched healthy individuals to evaluate the association between miR-146 and SLE’s ocular symptoms. Based on their results, patients with symptoms including dry eye, cataract, keratitis, and drusen showed less expression of miR-146 than asymptomatic patients. However, high expression of miR-146 was also shown in people with retinopathy symptoms. Consequently, a remarkable association was found between miR-146 and light sensitivity in SLE disease. 126 Of note, an association with secondary antiphospholipid syndrome (APS), which is positive in 30–40% of SLE patients, has been linked to miR-146. Even though there was no significant difference in miR-146 expression between patients with and without secondary APS, SLE patients had decreased expression of this miR-146 when compared with the control group. 123 Stimulation of SLE-associated renal injury in medical research laboratory lymphoproliferation (MRL/lpr) mice can be alleviated by miR-146a. 150 It was shown that miR-146a inhibited classical and nonclassical NF-κB pathways and moderated kidney damage by reducing auto-antibody production through B cells such as anti-dsDNA, anti-single-stranded DNA (anti-ssDNA), and anti-nuclear antibody (ANA). Furthermore, miR-146a can regulate the amount of protein and blood urea nitrogen (BUN) in the urine. Moreover, it can diminish inflammatory factors in the kidney tissues of MRL/lpr mice and moderate their effects on this organ. 150 Considering the obvious and non-negligible role of senescence of MSCs in SLE progression and decline of miR-146a expression in these patients, Dong and colleagues investigated the function of miR-146a in the control of MSC aging using the SA-gal assay. By imitating the functions of miR-146, they attempted to reverse the aging process in these patients, which shows the regulatory role of exosomal miR-146a in this pathway. 125 Also, according to Dominguez-Gutierrez et al., 124 the loss of the ability of miR-146 in SLE patients may reduce its efficiency in regulating the STAT1a isoform.

MiR-210

MiR-210 is an ncRNA that is often induced by hypoxia and is therefore called the ‘hypoxamir master’. 151 MiR-210 has a stem-loop on the intron of chromosome 11p15.5 regulated by both the hypoxia-inducible factor (HIF)-1α and HIF2α genes and is involved in tumor progression, myocardial infarction, and ischemic skin lesions with irregular expression.152154 According to several studies, miR-210 and HIF1α may be related to a variety of auto-immune diseases including SLE. By purifying the CD4+ T lymphocytes of lupus susceptible mice, high expression of miR-210 and HIF1α was shown on these cells. However, neither of these two factors was significantly altered in CD8+ T cells or CD19+ cells. Nevertheless, the miR-210/HIF1α pathway and its irregular expression can be a new regulatory pathway in SLE. 155 Similarly, in a similar study, miR-210 and HIF1, ERK1/2, and PI3 K/AKT were investigated, and it was indicated that the recognition of these biomolecules can play a part in the diagnosis of SLE. 113

MiR-21

MiR-21, an oncomiR, is known as a tumor suppressor due to its considerable role in apoptosis and necrosis and its importance in a wide variety of malignancies like breast, colon, and gastric cancer. 156 Remarkably, miR-21 has become significant in auto-immune diseases. Despite preserving its low expression in inactivated T lymphocytes, its expression rate is significantly increased after T-cell activation.157159 According to studies, miR-21 causes auto-immune disorders such as T1D, psoriasis, MS, RA, and SLE via stimulating inflammatory pathways. 156 MicroRNA profile identified miR-21 as an upregulated ncRNA in SLE CD4+ T lymphocytes. Moreover, further studies demonstrate that miR-21 expression is significantly enhanced in SLE patients. In addition, this upregulation was more pronounced in people with lower C3 complement levels than in others. 114 As hypoxia is one of the causes of miR-21 induction, it can affect the pathogenesis of SLE. The study of miR-21 in the serum, urine, and T cells of SLE patients showed more production of miR-21 in these patients than in healthy people. As a result, miR-21 might be utilized as a biomarker to diagnose and treat SLE. 113

MiR29a

This 64-base microRNA is an additional ncRNA located on the 7q32.3 chromosome. 160 MiR-29a is not only involved in functions such as cell proliferation, differentiation, apoptosis, angiogenesis, tumorigenesis, and metastasis but also by binding to the genes that stimulate or inhibit the progression of cancer has carcinogenicity in several neoplasms. 161 It seems that miR-29a, like other microRNAs, can be effective in auto-immune diseases. By comparing miR-29a in the groups of SLE patients with controls, a considerable decline in the amount of this microRNA was noticed in the B lymphocytes of the first group. Downregulation of miR-29a in SLE patients has been shown to enhance Crk-like protein expression, which in turn increases IgG secretion. Therefore, it is believed that adjusting the expression of miR-29a can improve the condition of SLE patients. 130

MiR-153-3p

Other noncoding microRNAs include miR-153-3p identified in human laryngeal squamous cell carcinoma and breast cancer. 162 Similarly, this ncRNA appears to be implicated in auto-immune diseases such as SLE. According to an investigation by Li et al., 163 miR-153-3p was shown to be elevated in SLE patients and was closely associated with the activity of the disease. Based on their results, miR-153-3p overexpression reduced UC-MSCs’ migration and proliferation as well as their ability to reduce Tfhs and enhance Tregs by suppressing PELI1. In addition, the upregulation of miR-153-3p diminished the favorable efficacy of UC-MSCs in MRL/lpr mice in vivo. 163 Hence, miR-153-3p can be considered a therapeutic target for treating SLE.

MiR-125b

MiR-125b-1 and miR-125b-2 are components of miR-125b involved in controlling various signaling pathways such as NF-κB, p53, PI3 K/Akt/mTOR, ErbB2 (erythroblastic oncogene B), and Wnt (Wingless-related integration site). In addition, they may have an essential role in cell proliferation, differentiation, apoptosis, drug resistance, and tumor survival. Increased expression of miR-125b has been revealed in auto-immune diseases such as psoriasis. 164 Given this issue, it is thought that alterations in its expression may also play a role in the pathogenesis of SLE. By studying the effect of UVB on the expression of miR-125-b, reduced values of miR-125-b were observed in both control and patient groups. However, its expression was significantly lower in SLE patients exposed to UVB than in normal individuals. Finally, downregulation in miR-125-b increased UV radiation resistance–associated gene (UVRAG) expression, which also increased autophagy in the PBMC of these patients. 128

MiR-199-3p

One of the microRNAs that appears to be implicated in cancer is miR-199a-3p. Decreased expression of this biomarker in malignancies such as kidney and bladder cancer has shown its role as a tumor suppressor. In addition, it can act as an oncogene with upregulation in gastric and colorectal cancers. 165 MiR-199-3p may have a role in the development of auto-immune disorders like SLE. According to studies with upregulated amounts of IL-10 and decreased amounts of PARP-1 expression in SLE patients, Xiaoping et al. examined the association between miR-199a-3p and these factors. It was demonstrated that the expression of miR-199-3p was increased in SLE patients. In this regard, activation of the ERK/2 pathway inhibited PARP-1 expression and increased IL-10 production in SLE patients. Considering the alterations related to miR-199-3p expression and its positive and negative correlation with IL-10 and PARP-1, respectively, restoring physiological miR-199-3p levels may be a remarkable breakthrough in the treatment of SLE. 131

Other microRNAs

Based on previous data on enhanced differentiation of Th17 cells in SLE by myeloid-derived suppressor cell (MDSC)–arginase (Arg)-1, Pang and colleagues used RNA chips to identify the microRNA regulatory network between MDSC and Th17 cells. They have indicated that increasing miR-322-5p expression not only increases the differentiation of Th17 and Treg but also activates the TGF-β pathway. Since miR-322-5p expression decreased with the Arg-1 inhibitor, the Arg-1/miR-322-5p axis can be a potential therapeutic target for SLE. 166 Furthermore, since Resolvin D1 (RvD1) is a suppressor of inflammatory responses, it has been linked to a Treg/Th17 imbalance in the pathogenesis of SLE. miR-30e-5p is involved in the Treg/Th17 differentiation as an RvD1 downstream microRNA. Thus, RvD1 may enhance SLE progression by upregulating Treg and reducing Th17 cell expression via miR-30e-5p. 167 Given the importance of IL-2 in immune tolerance, its inadequate production by epigenetic modifications such as miR-200a-3p could have an impact on SLE pathogenesis. Katsuyama et al. 127 have reported that decreased expression of miR-200a-3p subsequently reduced IL-2 levels by zinc finger E-box binding homeobox (ZEB)1 and C-terminal binding protein 2 (CtBP2), which highlights the crucial role of microRNA in this disease. As previously stated, pyroptosis and GSDMD are crucial in the pathophysiology of SLE. GSDMD can act as the ultimate pyroptosis tormentor in the activation of inflammatory responses and it may be a promising therapeutic target for a variety of disorders (NCT03984227). MiR-379-5p is located in the DLK1-DIO3 region, associated with organ growth and various diseases such as cancer. While GSDMD is the primary target of miR-379-5p, high levels of miR-379-5p expression have been shown to inhibit the increased effect of arsenite-induced GSDMD levels (NCT03984227).

The role of LncRNAs in the progression of SLE

LncRNAs are RNA molecules of at least 200 nt in length that are incapable of encoding proteins. Since the early 1990s, about 172,216 copies of the human species have been registered in the NONCODE database. 168 These RNAs are transcribed mostly by Pol II/Pol I RNA polymerase (Pol) and sometimes by Pol III RNA. 169 Despite the minor part of LncRNAs in protein coding, they have imperative functions in a variety of biological and physiopathological contexts. In line with this LncRNAs have the potential to engage in cellular organization and control, including DNA replication, RNA transcription, protein translation, cell development, and cell differentiation.170,171 LncRNAs can modify chromatin function, control the assembly and function of membraneless nuclear bodies, modify the stability and translation of cytoplasmic mRNAs, and interfere with signaling pathways depending on their localization and the specific interactions they have with DNA, RNA, and proteins. In diverse biological and physiopathological contexts, these functions ultimately affect gene expression. 12 Due to their exceptional function in regulating cellular processes, they also play a role in the advancement of a variety of disorders such as cancer, inflammation, and auto-immune disease.172,173 Due to alterations in LncRNA expression and its association with SLE in blood, PBMCs, T cells, monocyte-derived dendritic cells (moDCs), and plasma of patients compared with the control group, a significant correlation of LncRNA with SLE pathogenesis was found.174176 For instance, a clinical study evaluated the expression status of these LncRNAs in the blood samples of SLE patients and compared them with healthy individuals with real-time polymerase chain reaction (PCR) for 1 year (NCT03185767). There is evidence to suggest a link between LncRNA and how they interfere with SLE development, as outlined below. Figure 3 and Table 2 illustrate the association between LncRNAs and SLE.

Figure 3.

Figure 3.

LncRNAs associated with SLE pathogenesis. Like microRNAs, a substantial number of LncRNAs have been acknowledged that can influence the pathogenesis of SLE. Also, the effect of different SNPs of these LncRNAs on the onset of SLE symptoms can be less effective. The impact of the LncRNAs network and their association with the MAPK pathway has also identified them as diagnostic biomarkers in SLE.

C, complement; CRP, C-reactive protein; E4BP4, transcriptional repressor E4-binding protein 4; GAS5, growth arrest–specific 5; IL, interleukins; MALAT-1, metastasis-associated lung adenocarcinoma transcript 1; MAPK, microtubule-associated protein kinase; NEAT1, nuclear enriched abundant transcript 1; NF-κB, nuclear factor-kappa B; PBMC, peripheral blood mononuclear cell; SIRT1, sirtuin 1; SLE, systemic lupus erythematosus; T helper, Th; TUG1, taurine-upregulated gene 1.

Table 2.

LncRNAs related to SLE disease.

LncRNA Number of clinical samples Biological sample Target gene Result References
NEAT-1 97 SLE patients, 50 healthy group PBMC Th1/Th2 NEAT1 is upregulated in SLE PBMCs relative to controls. 177
29 SLE patients and 40 age- and gender-matched normal controls PBMC IL-6, CXCL10 NEAT1 expression was higher in SLE patients compared with controls. 178
TUG1 85 patients with SLE, 95 healthy controls PBMC Complement C3 TUG1 LncRNA levels were decreased in SLE patients. 179
24 SPF BALB/c female mice Serum Nuclear factor-kappa B (NF-κB) Among all groups, the SLE group showed the highest levels of TUG1 and p65 expression in the cytoplasm. 180
GAS5 302 SLE patients, 396 healthy control PBMC PTEN In comparison with healthy volunteers, SLE patients showed substantially lower amounts of LncRNA-GAS5 in their PBMCs. 181
30 patients with SLE, 20 healthy controls CD4+ T cell E4BP4, miR-92a-3p GAS5 levels were lower in SLE patients’ CD4+ T cells. 182
45 SLE patients, 45 SLE patients CD4+ T cells Complement component 3 (C3) GAS5 levels in CD4+ T cells from SLE patients were substantially higher than those in control participants. 114
187 patients with SLE, 8 healthy controls Plasma ESR GAS5 expression was shown to be significantly reduced in SLE patients. 183
85 SLE patients and 71 healthy controls PBMC C-reactive protein (CRP) In SLE patients, GAS5 expression was considerably lower than in healthy controls. 184
linc0597 187 patients with SLE, 8 healthy controls Plasma C3 In SLE patients, linc0597 was significantly overexpressed. 183
85 SLE patients and 71 healthy controls PBMC _ Linc0597 expression levels in SLE patients were considerably lower than in healthy controls. 184
Lnc-DC 187 patients with SLE, 8 healthy controls Plasma C3 No significant differences 183
85 SLE patients and 71 healthy controls PBMC C-reactive protein (CRP) Lnc-DC expression levels were significantly lower in SLE patients than in healthy controls. 184
linc0949 187 patients with SLE, 8 healthy controls Plasma Anti-dsDNA No significant differences 183
85 SLE patients and 71 healthy controls PBMC _ Between SLE patients and healthy controls, there were no significant variations in linc0949 levels. 184
Lnc0640 76 SLE patients and 71 controls PBMC _ 185
Lnc3643 76 SLE patients and 71 controls PBMC CRP, ESR In SLE patients with proteinuria, Lnc3643 levels were statistically substantially lower than in those without. 185
Lnc5150 76 SLE patients and 71 controls PBMC ESR The levels of Lnc5150 expression were statistically substantially lower. 185
Lnc7514 76 SLE patients and 71 controls PBMC SLEDAI-2 K, ESR In anti-dsDNA+ patients, nc7514 levels were statistically substantially lower than in anti-dsDNA (-) patients. 185
Lncagf 76 SLE patients and 71 controls PBMC _ 185
MALAT-1 36 SLE patients and 45 healthy controls PBMCs and monocytes IL-21 and SIRT1 In SLE, MALAT-1 expression was unusually high. 186

Anti-dsDNA, anti-double-stranded DNA; BALB/C, Bagg Albino; C, complement; CRP, C-reactive protein; CXCL10, C-X-C motif chemokine ligand 10; E4BP4, transcriptional repressor E4-binding protein 4; ESR, erythrocyte sedimentation rate; GAS5, growth arrest–specific 5; IL, interleukins; MALAT-1, metastasis-associated lung adenocarcinoma transcript 1; NEAT1, nuclear enriched abundant transcript 1; NF-κB, nuclear factor-kappa B; PBMC, Peripheral blood mononuclear cell; SIRT1, sirtuin 1; SLE, systemic lupus erythematosus; SLEDAI, Systemic Lupus Erythematosus Disease Activity Index; T helper, Th; TUG1, taurine-upregulated gene 1.

Nuclear paraspeckle assembly text 1

Nuclear paraspeckle assembly text 1 (NEAT1) is 3.2 kb ncRNA located on the 11q13.1 chromosome in the LncRNA subtypes. This LncRNA is produced by transcription from the familial tumor syndrome multiple endocrine neoplasia (MEN) type 1. Two isoforms, i.e. NEAT1-1 and NEAT1-2, have been discovered from this ncRNA that alternative 3′-end processing is responsible for regulating them. 187 Due to its high expression in non-neural cells and cell lines, it participates in several disorders, including cancers like non–small cell lung cancer, ovarian cancer, cervical cancer, hepatocellular carcinoma, etc. 188 Numerous studies have shown an association between NEAT-1 and SLE. 189 Jiang et al. evaluated the expression of NEAT1 in PBMC from patients with SLE and healthy individuals. NEAT1 was shown to be highly upregulated in the PBMC of SLE patients in this research. Consistent with this, a negative connection between its expression and Th1/Th2 balance was reported. Because the Th1/Th2 balance is crucial for the immune system to work correctly, altering it causes abnormal immune responses associated with SLE development. 177 NEAT-1 may have a role in lupus pathogenesis by modulating inflammatory pathways via MAPK. In this regard, individuals with SLE exhibited high levels of NEAT-1, mostly generated by monocytes, compared with controls. They can enhance SLE activity by increasing the number of monocytes in PBMC and the excessive production of inflammatory mediators such as cytokines and chemokines as a result of NEAT-1 overexpression. 178 Also, based on the findings of Dong et al. regarding the correlation between increased NEAT1 expression and granulocytic MDSCs (G-MDSC) in MRL/lpr mice in SLE, they have indicated that NEAT-1 upregulation activates the IFN-I signaling pathway of B cells through BAFF and promotes G-MDSCs, which cause SLE progression. 190

Taurine-upregulated gene 1

Taurine-upregulated gene 1 (TUG1) generates LncRNAs that interact with the polycomb repressor complex to regulate transcription epigenetically. This gene, located on chromosome 22q12.2, promotes cell proliferation and has been linked to cancers such as liver, osteosarcoma, glioma, and bladder. 191 TUG1 has a protective effect being linked to the NF-κB pathway because it can inhibit apoptosis and the production of inflammatory elements.192,193 Cao et al. performed pyrrolidine dithiocarbamate (PTDC) therapy to demonstrate the influence of NF-κB inhibition on renal damage in the SLE mouse model and also its association with TUG1. According to the results, treatment with PTDC increased TUG1 expression and downregulated P65 NF-κB. In brief, this highlights the negative association between TUG1 and NF-B p65 as the effect of these LncRNAs on SLE. 180 They also noted the downregulation of TUG1 expression in SLE patients’ PBMC compared with healthy individuals. 179

Growth arrest–specific 5

The gene encoding growth arrest–specific 5 (GAS5) is found on the 1q25.1 chromosome, which also contains numerous C/D box snoRNA (small nucleolar RNAs) genes in its introns. By interacting with the DNA-binding domain of the glucocorticoid receptor (GCR), this gene can imitate the glucocorticoid response. As a result, it plays a critical function in the androgen receptor, progesterone, and mineralocorticoid regulation. GAS5 may act as a tumor suppressor in diagnosis and management due to its role in suppressing proliferation and cell death. 194 GAS5 is implicated in malignancies and inflammatory illnesses, but its action on GCRs has also been linked to auto-immune disorders. Several GAS5 SNPs have been discovered, including rs145204276, rs2235095, rs6790, rs2067079, and rs1951625, which may be linked to SLE. The effect of the rs145204276 polymorphism on GAS5 expression in SLE patients is a notable example. Individuals with the rs145204276 ID+ genotype had more GAS5 expression than those without it, and they had fewer SLE symptoms, indicating that this polymorphism may be protective. 181 Qian and coworkers discovered a correlation between GAS5 expression changes and SLE based on previous investigations of GAS5 gene localization information. GAS5 was also shown to be downregulated in SLE patients’ T cells and plasma. GAS5 overexpression also inhibits miR-92a-3p, which controls adenovirus E4-binding protein 4 (E4BP4). As a result, it is critical in suppressing T lymphocyte activation in SLE patients. 182 However, during experiments on GAS5 alteration levels in SLE patients, Wu et al. 183 demonstrated a remarkable reduction in its expression in these patients compared with controls. Suo and colleagues surveyed the association of GAS5 with CD4+ T lymphocytes. Due to the increasing trend of GAS5 in patients with pleurisy, rash, anti-dsDNA, and low complement C3, it can be considered for the identification of SLE. In contrast to previous studies, this investigation emphasizes the high level of GAS in CD4+ T cells in SLE patients compared with healthy subjects. 114 Wu et al. 195 examined and analyzed ncRNAs, including GAS5, lnc0640, lnc3643, lnc6655, and lnc7074, as a competing endogenous RNA network (ceRNA). They also analyzed the association of GAS5, lnc0640, lnc3643, lnc6655, and lnc7074 with the pathogenesis of SLE through the inhibition of microRNAs. According to their results, these LncRNAs, which act through the MAPK pathway, might be utilized as novel and diagnostic biomarkers in SLE. 195

Lnc-DC

Lnc-DC is one of the most discussed ncRNA types expressed in DCs. Lnc-DC is capable of maturing DCs via STAT3 phosphorylation. In addition, the impact of Lnc-DC on monocytes causing them to differentiate into DCs can induce T lymphocyte activation. 196 Currently, Lnc-DC has become a central issue in the field of auto-immune disorders such as Sjögren, MS, and SLE. 197 In SLE, Lnc-DC has been identified as a significant biomarker. By examining this factor in SLE patients and normal individuals, Wu et al. 183 achieved a decrease in its expression in lupus patients. Li and colleagues investigated the expression levels of linc0597 Lnc-DC and GAS5 in SLE patients and found a reduction in their expression in SLE. It is worth mentioning that a change in the number of polymorphisms in the mentioned LncRNAs is not related to an enhanced risk of this disorder. 184

Myocardial infarction–associated transcript

Myocardial infarction–associated transcript (MIAT) is a common part of LncRNAs first identified in 2006 as the myocardial infarction gene. MIAT has a wide range of applications in cancer and another disease such as diabetic retinopathy, paranoid schizophrenia, and vascular dysfunction. 198 Even though MIAT is poorly known, it appears to play a critical role in auto-immune disorders. 199 Xu et al. explored the vast network between LncRNA, microRNA, and mRNA called ceRNA. They aimed to find an association between differentially expressed LncRNAs (DE-LncRNAs) in SLE pathogenesis. Interestingly, the influence of particular DE-LncRNAs like MIAT and NEAT1 by binding to mRNA and inhibiting the expression of the corresponding miRNA was observed in SLE. Consequently, LncRNAs, in addition to having a direct effect on SLE disease, also play a pivotal part in the disease by the ceRNA network. 200 Furthermore, investigations on MRL/lpr mice showed that MIAT exacerbates the progression of SLE. Hematoxylin and eosin (H&E) staining of the kidney tissues of MRL/lpr mice showed that MIAT, as a competitive inhibitor of miR-222, by increasing CFHR5 expression, degrades miR-222 and enhances SLE activity. 201

Metastasis-associated lung adenocarcinoma transcript-1

Metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1), with a size of 12,820 bp, is an RNA gene affiliated with the LncRNA class. Principal physiological fundamental characteristics of MALAT-1 are alternative splicing and epigenetic changes. 202 However, it is pathologically involved in various diseases such as hyperglycemia and vulva squamous cell carcinoma. 203 MALAT-1 has recently been discovered to be involved in SLE as a novel inflammatory regulator. Increasing the unusual expression of this biomarker in SLE, especially in monocytes, and the destructive effects of MALAT-1 by the Sirt1 pathway, display its central role in the pathogenesis of SLE. MALAT-1 greatly increased IL-2 secretion in SLE patients compared with healthy individuals. 186

Other LncRNAs

There are additional ncRNAs that have not been investigated sufficiently but may have an impact on SLE. Wang et al. studied the expression pattern of five LncRNAs in the PBMC of SLE patients compared with the controls in the research. The abnormal expression of Lnc3643 and Lnc7514 was related to C-reactive protein (CRP) and positive anti-dsDNA, respectively. Accordingly, Lnc7514 expression in SLE patients was decreased with positive anti-dsDNA patients compared with negative anti-dsDNA. The analysis also revealed a correlation between CRP and the level of expression for Lnc3643. 185

The role of circRNAs in the progression of SLE

CircRNAs are a vast family of ncRNAs generated by reverse splicing of pre-mRNAs; meanwhile, their expression is also influenced by pre-mRNA transcription levels, which are determined by transcription factor activity and epigenetic modifications such as methylation and acetylation. 204 They have a unique structure consisting of a continuous covalently closed loop that lacks a 5′-cap and a 3′-poly A tail. The specific characteristics of circRNAs have made them resistant to degradation by exonuclease ribonucleases. 204 Viroids are one of the first circRNA molecules found in the last 40 years, with the exception that they are not produced by the reverse splicing mechanism. 205 Moreover, circRNAs are often synthesized in the nucleus, although they can also be detected in the cytoplasm and in the nucleus.206,207 Despite electron microscope observations of circRNAs in the cytoplasm of eukaryotic cells, they were regarded as ‘junk’ generated by improper splicing processes. 204 Considering the abundance of circRNAs in body fluids, recent studies have pointed to their enrichment and transport by exosomes. Therefore, the secretion of exosomes from pathological cells can cause an abnormal increase of certain circRNAs, which shows their importance as valuable biomarkers.208210 A considerable number of studies have revealed that circRNAs have a role in a variety of disorders such as auto-immune diseases, indicating their potential as clinical biomarkers and therapeutic targets. 211 So far, using microarray and high-throughput sequencing techniques, several aberrantly expressed circRNAs have been identified in SLE. 212 Luo et al. 213 investigated the level of peripheral blood circRNAs in SLE patients with renal involvement (SLE + RI) and indicated that the level of hsa-circ-0082688 and hsa-circ-0008675 in SLE + RI patients was enhanced. Furthermore, new research in circRNA’s context has revealed that these RNAs absorb miRNAs like sponges and influence downstream target genes. The elevated expression of hsa-circ-0002003 in SLE may be involved in controlling immunological responses, according to the investigation of Lin et al. 214 On the other hand, Zhang et al. attempted to examine the regulation mechanism of circRNAs in T cells from SLE patients and revealed a correlation between the circRNA–microRNA–mRNA regulatory network. They established a circRNA–microRNA–miRNA network of 8 differentially expressed circRNAs (DECs), 4 miRNAs, and 13 mRNAs using several bioinformatics. Furthermore, they noted that their study may improve the diagnosis, treatment, and prognosis of SLE patients. 215 Zheng et al. stated that they extracted circRNAs for detection from SLE patients’ PBMCs. They discovered the aberrant expression of certain circRNAs in PBMCs. Thus, circRNAs may be beneficial as unique biomarkers for the diagnosis of SLE. 216 Considering the importance of circRNAs in the diagnosis of SLE, Miao et al. investigated 128 circRNAs in PBMCs of patients. According to the study’s findings, a reduction in circPTPN22 in patients with greater SLEDAI revealed a negative connection between these two variables. 217 Furthermore, another study demonstrated the function of hsa_circ_0045272 in the negative regulation of apoptosis and IL-2 secretion in SLE. 218 In addition, Zhang et al. 219 reported reduced expression of the hsa_circRNA_407176 and hsa_circRNA_001308 in the plasma and PBMCs of SLE patients. Generally, circRNAs can behave as novel biomarkers and essential regulators of SLE pathogenesis. However, information on the intricacies of the circRNA mechanism in SLE is inadequate.

Conclusion

The development of an immune response in the absence of risk factors might result in diseases such as asthma, allergies, or auto-immune diseases. In auto-immune disorders, the body mistakenly attacks the body’s own tissues. It has been shown that factors such as microorganisms and drugs may cause changes in the immune system in genetically predisposed individuals. Furthermore, these disorders can be influenced by a variety of factors such as inheritance, environment, hormones, and infectious agents; yet, despite the availability of many medications, no definite cure for this ailment has been established. SLE is a chronic disease with a wide variety of medical symptoms caused by complicated mechanisms such as incorrect apoptosis, dysregulation of the innate and adaptive immune systems, and complement activation. The pattern of clinical presentations, on the other hand, differs from person to person according to heterogeneity, emphasizing the relevance of genetic and environmental variables in this disease. Methylation, histone modification, and, in particular, ncRNAs are all factors in the pathogenesis of SLE. Despite their lack of participation in protein synthesis, ncRNAs play a crucial function in controlling gene expression due to their formation of a major portion of the human genome. Due to the significant role of ncRNAs in B-cell activation pathways and NF-κB pathways, as well as STAT-related pathways in SLE pathogenesis, new therapeutic goals have emerged as a result of research. In addition, greater attention should be paid to the interaction between microRNAs, LncRNAs, and circRNAs in this disorder, and more targeted therapies should be explored in this context. ncRNAs, particularly microRNAs, have been suggested to play a role in the identification of SLE complications, in addition to detecting healthy people from patients.

Acknowledgments

Not applicable.

Footnotes

ORCID iD: Behzad Baradaran Inline graphichttps://orcid.org/0000-0002-8642-6795

Contributor Information

Mina Afrashteh Nour, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.

Farid Ghorbaninezhad, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.

Zahra Asadzadeh, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.

Amir Baghbanzadeh, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.

Hamidreza Hassanian, Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.

Patrizia Leone, Department of Interdisciplinary Medicine, University of Bari ‘Aldo Moro’, Bari, Italy.

Mahdi Jafarlou, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.

Nazila Alizadeh, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.

Vito Racanelli, Department of Interdisciplinary Medicine, University of Bari ‘Aldo Moro’, 70124, Bari, Italy.

Behzad Baradaran, Immunology Research Center, Tabriz University of Medical Sciences, Daneshgah Street, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.

Declarations

Ethics approval and consent to participate: Not applicable.

Consent for publication: Not applicable.

Author contributions: Mina Afrashteh Nour: Conceptualization; Investigation; Writing – original draft.

Farid Ghorbaninezhad: Conceptualization; Investigation; Writing – original draft.

Zahra Asadzadeh: Conceptualization; Data curation; Methodology; Software; Visualization.

Amir Baghbanzadeh: Formal analysis; Investigation; Software; Visualization.

Hamidreza Hassanian: Investigation; Methodology; Software; Visualization.

Patrizia Leone: Investigation; Methodology; Software; Visualization.

Mahdi Jafarlou: Methodology; Validation; Writing – review & editing.

Nazila Alizadeh: Formal analysis; Validation; Writing – review & editing.

Vito Racanelli: Formal analysis; Supervision; Validation; Writing – review & editing.

Behzad Baradaran: Formal analysis; Supervision; Validation; Writing – review & editing.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Availability of data and materials: Not applicable.

References

  • 1.Smith DA, Germolec DR.Introduction to immunology and autoimmunity. Environ Health Perspect 1999; 107(Suppl. 5): 661–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rosenblum MD, Remedios KA, Abbas AK.Mechanisms of human autoimmunity. J Clin Invest 2015; 125: 2228–2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Firestein GS, Gabriel SE, McInnes IB, et al. Kelley and Firestein’s textbook of rheumatology. Elsevier Health Sciences, 2017. [Google Scholar]
  • 4.Firestein GS, Budd RC, Gabriel SE, et al. Kelley and Firestein’s textbook of rheumatology. Elsevier Health Sciences, 2016. [Google Scholar]
  • 5.Ali A, Sayyed Z, Ameer MA, et al. Systemic lupus erythematosus: an overview of the disease pathology and its management. Cureus 2018; 10: e3288. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 6.Borchers AT, Naguwa SM, Shoenfeld Y, et al. The geoepidemiology of systemic lupus erythematosus. Autoimmun Rev 2010; 9: A277–A287. [DOI] [PubMed] [Google Scholar]
  • 7.Winkle M, El-Daly SM, Fabbri M, et al. Noncoding RNA therapeutics – challenges and potential solutions. Nat Rev Drug Discov 2021; 20: 629–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Si W, Shen J, Zheng H, et al. The role and mechanisms of action of microRNAs in cancer drug resistance. Clin Epigenetics 2019; 11: 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wahid F, Shehzad A, Khan T, et al. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta 2010; 1803: 1231–1243. [DOI] [PubMed] [Google Scholar]
  • 10.Giovannetti E, Erozenci A, Smit J, et al. Molecular mechanisms underlying the role of microRNAs (miRNAs) in anticancer drug resistance and implications for clinical practice. Crit Rev Oncol Hematol 2012; 81: 103–122. [DOI] [PubMed] [Google Scholar]
  • 11.Jiangpan P, Qingsheng M, Zhiwen Y, et al. Emerging role of microRNA in neuropathic pain. Curr Drug Metab 2016; 17: 336–344. [DOI] [PubMed] [Google Scholar]
  • 12.Statello L, Guo C-J, Chen L-L, et al. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol 2021; 22: 96–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Szabo L, Salzman J.Detecting circular RNAs: bioinformatic and experimental challenges. Nat Rev Genet 2016; 17: 679–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Taheri M, Eghtedarian R, Dinger ME, et al. Exploring the role of non-coding RNAs in the pathophysiology of systemic lupus erythematosus. Biomolecules 2020; 10: 937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cooper GS, Stroehla BC.The epidemiology of autoimmune diseases. Autoimmun Rev 2003; 2: 119–125. [DOI] [PubMed] [Google Scholar]
  • 16.Senousy MA, Shaker OG, Sayed NH, et al. LncRNA GAS5 and miR-137 polymorphisms and expression are associated with multiple sclerosis risk: mechanistic insights and potential clinical impact. ACS Chem Neurosci 2020; 11: 1651–1660. [DOI] [PubMed] [Google Scholar]
  • 17.Chang C.Chapter 17 – managing autoimmune disorders through personalized epigenetic approaches. In: Tollefsbol TO. (ed.) Personalized epigenetics. Boston, MA: Academic Press, 2015, pp. 475–505. [Google Scholar]
  • 18.Wang L, Wang FS, Gershwin ME.Human autoimmune diseases: a comprehensive update. J Intern Med 2015; 278: 369–395. [DOI] [PubMed] [Google Scholar]
  • 19.Salinas GF, Braza F, Brouard S, et al. The role of B lymphocytes in the progression from autoimmunity to autoimmune disease. Clin Immunol 2013; 146: 34–45. [DOI] [PubMed] [Google Scholar]
  • 20.Panda S, Ding JL.Natural antibodies bridge innate and adaptive immunity. J Immunol 2015; 194: 13–20. [DOI] [PubMed] [Google Scholar]
  • 21.Pillai S.Rethinking mechanisms of autoimmune pathogenesis. J Autoimmun 2013; 45: 97–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schmidt CW.Questions persist: environmental factors in autoimmune disease. Environ Health Perspect 2011; 119: A249–A253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Marson A, Housley WJ, Hafler DA.Genetic basis of autoimmunity. J Clin Invest 2015; 125: 2234–2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Goris A, Liston A.The immunogenetic architecture of autoimmune disease. Cold Spring Harb Perspect Biol 2012; 4: a007260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sollid LM, Pos W, Wucherpfennig KW.Molecular mechanisms for contribution of MHC molecules to autoimmune diseases. Curr Opin Immunol 2014; 31: 24–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vandenbroeck K.Cytokine gene polymorphisms and human autoimmune disease in the era of genome-wide association studies. J Interferon Cytokine Res 2012; 32: 139–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bennett CL, Christie J, Ramsdell F, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001; 27: 20–21. [DOI] [PubMed] [Google Scholar]
  • 28.An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet 1997; 17: 399–403. [DOI] [PubMed] [Google Scholar]
  • 29.Colafrancesco S, Perricone C, Priori R, et al. Sjögren’s syndrome: another facet of the autoimmune/inflammatory syndrome induced by adjuvants (ASIA). J Autoimmun 2014; 51: 10–16. [DOI] [PubMed] [Google Scholar]
  • 30.Kivity S, Arango MT, Ehrenfeld M, et al. Infection and autoimmunity in Sjogren’s syndrome: a clinical study and comprehensive review. J Autoimmun 2014; 51: 17–22. [DOI] [PubMed] [Google Scholar]
  • 31.Peng J, Narasimhan S, Marchesi JR, et al. Long term effect of gut microbiota transfer on diabetes development. J Autoimmun 2014; 53: 85–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mills KH.TLR-dependent T cell activation in autoimmunity. Nat Rev Immunol 2011; 11: 807–822. [DOI] [PubMed] [Google Scholar]
  • 33.Root-Bernstein R, Fairweather D.Complexities in the relationship between infection and autoimmunity. Curr Allergy Asthma Rep 2014; 14: 407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Serafini B, Rosicarelli B, Franciotta D, et al. Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain. J Exp Med 2007; 204: 2899–2912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mikuls TR, Payne JB, Yu F, et al. Periodontitis and porphyromonas gingivalis in patients with rheumatoid arthritis. Arthrit Rheumatol 2014; 66: 1090–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mathis D, Benoist C.Microbiota and autoimmune disease: the hosted self. Cell Host Microbe 2011; 10: 297–301. [DOI] [PubMed] [Google Scholar]
  • 37.Kuhn A, Wenzel J, Weyd H.Photosensitivity, apoptosis, and cytokines in the pathogenesis of lupus erythematosus: a critical review. Clin Rev Allergy Immunol 2014; 47: 148–162. [DOI] [PubMed] [Google Scholar]
  • 38.Steinman RM, Turley S, Mellman I, et al. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med 2000; 191: 411–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bluestone JA, Tang Q, Sedwick CE.T regulatory cells in autoimmune diabetes: past challenges, future prospects. J Clin Immunol 2008; 28: 677–684. [DOI] [PubMed] [Google Scholar]
  • 40.Mohan JF, Unanue ER.Unconventional recognition of peptides by T cells and the implications for autoimmunity. Nat Rev Immunol 2012; 12: 721–728. [DOI] [PubMed] [Google Scholar]
  • 41.Mohan JF, Petzold SJ, Unanue ER.Register shifting of an insulin peptide-MHC complex allows diabetogenic T cells to escape thymic deletion. J Exp Med 2011; 208: 2375–2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mohan JF, Levisetti MG, Calderon B, et al. Unique autoreactive T cells recognize insulin peptides generated within the islets of Langerhans in autoimmune diabetes. Nat Immunol 2010; 11: 350–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kawasaki T, Kawai T, Akira S.Recognition of nucleic acids by pattern-recognition receptors and its relevance in autoimmunity. Immunol Rev 2011; 243: 61–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Turer EE, Tavares RM, Mortier E, et al. Homeostatic MyD88-dependent signals cause lethal inflammation in the absence of A20. J Exp Med 2008; 205: 451–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pisitkun P, Deane JA, Difilippantonio MJ, et al. Autoreactive B cell responses to RNA-related antigens due to TLR7 gene duplication. Science 2006; 312: 1669–1672. [DOI] [PubMed] [Google Scholar]
  • 46.Fortuna G, Brennan MT.Systemic lupus erythematosus: epidemiology, pathophysiology, manifestations, and management. Dental Clin 2013; 57: 631–655. [DOI] [PubMed] [Google Scholar]
  • 47.Blotzer JW.Systemic lupus erythematosus I: historical aspects. Md State Med J 1983; 32: 439–441. [PubMed] [Google Scholar]
  • 48.Smith CD, Cyr M.The history of lupus erythematosus. From Hippocrates to Osler. Rheum Dis Clin North Am 1988; 14: 1–14. [PubMed] [Google Scholar]
  • 49.Mok C, Lau C.Pathogenesis of systemic lupus erythematosus. J Clin Pathol 2003; 56: 481–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Systemic lupus erythematosus. Ann Int Med 2020; 172: ITC81–ITC96. [DOI] [PubMed] [Google Scholar]
  • 51.Borchers AT, Keen CL, Shoenfeld Y, et al. Surviving the butterfly and the wolf: mortality trends in systemic lupus erythematosus. Autoimmun Rev 2004; 3: 423–453. [DOI] [PubMed] [Google Scholar]
  • 52.Urowitz MB, Gladman DD, Tom BD, et al. Changing patterns in mortality and disease outcomes for patients with systemic lupus erythematosus. J Rheumatol 2008; 35: 2152–2158. [DOI] [PubMed] [Google Scholar]
  • 53.Bernatsky S, Boivin JF, Joseph L, et al. Mortality in systemic lupus erythematosus. Arthrit Rheumat 2006; 54: 2550–2557. [DOI] [PubMed] [Google Scholar]
  • 54.Kasitanon N, Magder LS, Petri M.Predictors of survival in systemic lupus erythematosus. Medicine 2006; 85: 147–156. [DOI] [PubMed] [Google Scholar]
  • 55.Yurasov S, Wardemann H, Hammersen J, et al. Defective B cell tolerance checkpoints in systemic lupus erythematosus. J Exp Med 2005; 201: 703–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Casciola-Rosen LA, Anhalt G, Rosen A.Autoantigens targeted in systemic lupus erythematosus are clustered in two populations of surface structures on apoptotic keratinocytes. J Exp Med 1994; 179: 1317–1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tsokos GC, Lo MS, Costa Reis P, et al. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2016; 12: 716–730. [DOI] [PubMed] [Google Scholar]
  • 58.Thanou A, Jupe E, Purushothaman M, et al. Clinical disease activity and flare in SLE: current concepts and novel biomarkers. J Autoimmun 2021; 119: 102615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Theofilopoulos AN, Kono DH, Beutler B, et al. Intracellular nucleic acid sensors and autoimmunity. J Interferon Cytokine Res 2011; 31: 867–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Deretic V, Saitoh T, Akira S.Autophagy in infection, inflammation and immunity. Nat Rev Immunol 2013; 13: 722–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Mahajan A, Herrmann M, Muñoz LE.Clearance deficiency and cell death pathways: a model for the pathogenesis of SLE. Front Immunol 2016; 7: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zan H, Tat C, Casali P.MicroRNAs in lupus. Autoimmunity 2014; 47: 272–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lu R, Munroe ME, Guthridge JM, et al. Dysregulation of innate and adaptive serum mediators precedes systemic lupus erythematosus classification and improves prognostic accuracy of autoantibodies. J Autoimmun 2016; 74: 182–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ohtsuka K, Gray JD, Stimmler MM, et al. The relationship between defects in lymphocyte production of transforming growth factor-beta1 in systemic lupus erythematosus and disease activity or severity. Lupus 1999; 8: 90–94. [DOI] [PubMed] [Google Scholar]
  • 65.Talaat RM, Mohamed SF, Bassyouni IH, et al. Th1/Th2/Th17/Treg cytokine imbalance in systemic lupus erythematosus (SLE) patients: correlation with disease activity. Cytokine 2015; 72: 146–153. [DOI] [PubMed] [Google Scholar]
  • 66.Yap DY, Lai KN.Pathogenesis of renal disease in systemic lupus erythematosus – the role of autoantibodies and lymphocytes subset abnormalities. Int J Mol Sci 2015; 16: 7917–7931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Harley IT, Kaufman KM, Langefeld CD, et al. Genetic susceptibility to SLE: new insights from fine mapping and genome-wide association studies. Nat Rev Genet 2009; 10: 285–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Belot A, Cimaz R.Monogenic forms of systemic lupus erythematosus: new insights into SLE pathogenesis. Pediatr Rheum Online J 2012; 10: 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Achtman JC, Werth VP.Pathophysiology of cutaneous lupus erythematosus. Arthr Res Ther 2015; 17: 182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Grimaldi CM.Sex and systemic lupus erythematosus: the role of the sex hormones estrogen and prolactin on the regulation of autoreactive B cells. Curr Opin Rheumatol 2006; 18: 456–461. [DOI] [PubMed] [Google Scholar]
  • 71.Shelly S, Boaz M, Orbach H.Prolactin and autoimmunity. Autoimmun Rev 2012; 11: A465–470. [DOI] [PubMed] [Google Scholar]
  • 72.Cunningham M, Gilkeson G.Estrogen receptors in immunity and autoimmunity. Clin Rev Allergy Immunol 2011; 40: 66–73. [DOI] [PubMed] [Google Scholar]
  • 73.Barbhaiya M, Costenbader KH.Ultraviolet radiation and systemic lupus erythematosus. Lupus 2014; 23: 588–595. [DOI] [PubMed] [Google Scholar]
  • 74.Nelson P, Rylance P, Roden D, et al. Viruses as potential pathogenic agents in systemic lupus erythematosus. Lupus 2014; 23: 596–605. [DOI] [PubMed] [Google Scholar]
  • 75.Fang K, Orr-Skirvin JA.Systemic lupus erythematosus, its treatment, and future directions. US Pharm 2019; 44: 3–7. [Google Scholar]
  • 76.Ferretti C, La Cava A. Overview of the pathogenesis of systemic lupus erythematosus. In: Tsokos GC. (ed.) Systemic lupus erythematosus. Amsterdam: Elsevier, 2016, pp. 55–62. [Google Scholar]
  • 77.Catalina MD, Owen KA, Labonte AC, et al. The pathogenesis of systemic lupus erythematosus: harnessing big data to understand the molecular basis of lupus. J Autoimmun 2020; 110: 102359. [DOI] [PubMed] [Google Scholar]
  • 78.Shirai T, Hirose S.Molecular pathogenesis of SLE. Springer Semin Immunopathol 2006; 28: 79–82. [DOI] [PubMed] [Google Scholar]
  • 79.Moulton VR, Suarez-Fueyo A, Meidan E, et al. Pathogenesis of human systemic lupus erythematosus: a cellular perspective. Trends Mol Med 2017; 23: 615–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Harley JB, Alarcón-Riquelme ME, Criswell LA, et al. Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet 2008; 40: 204–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ghodke-Puranik Y, Niewold TB.Immunogenetics of systemic lupus erythematosus: a comprehensive review. J Autoimmun 2015; 64: 125–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Bentham J, Morris DL, Graham DSC, et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat Genet 2015; 47: 1457–1464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Langefeld CD, Ainsworth HC, Cunninghame Graham DS, et al. Transancestral mapping and genetic load in systemic lupus erythematosus. Nat Commun 2017; 8: 16021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bluestone JA, Bour-Jordan H, Cheng M, et al. T cells in the control of organ-specific autoimmunity. J Clin Invest 2015; 125: 2250–2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Verthelyi D, Ahmed SA.17 beta-estradiol, but not 5 alpha-dihydrotestosterone, augments antibodies to double-stranded deoxyribonucleic acid in nonautoimmune C57BL/6J mice. Endocrinology 1994; 135: 2615–2622. [DOI] [PubMed] [Google Scholar]
  • 86.Grimaldi CM, Cleary J, Dagtas AS, et al. Estrogen alters thresholds for B cell apoptosis and activation. J Clin Invest 2002; 109: 1625–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pathak S, Mohan C.Cellular and molecular pathogenesis of systemic lupus erythematosus: lessons from animal models. Arthritis Res Ther 2011; 13: 241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Sawalha AH, Jeffries M, Webb R, et al. Defective T-cell ERK signaling induces interferon-regulated gene expression and overexpression of methylation-sensitive genes similar to lupus patients. Genes Immun 2008; 9: 368–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Coit P, Renauer P, Jeffries MA, et al. Renal involvement in lupus is characterized by unique DNA methylation changes in naïve CD4+ T cells. J Autoimmun 2015; 61: 29–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gorelik G, Richardson B.Key role of ERK pathway signaling in lupus. Autoimmunity 2010; 43: 17–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Deng C, Kaplan MJ, Yang J, et al. Decreased Ras-mitogen-activated protein kinase signaling may cause DNA hypomethylation in T lymphocytes from lupus patients. Arthritis Rheum 2001; 44: 397–407. [DOI] [PubMed] [Google Scholar]
  • 92.Swaak AJ, Groenwold J, Aarden LA, et al. Prognostic value of anti-dsDNA in SLE. Ann Rheum Dis 1982; 41: 388–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mi XB, Zeng FQ.Hypomethylation of interleukin-4 and -6 promoters in T cells from systemic lupus erythematosus patients. Acta Pharmacol Sin 2008; 29: 105–112. [DOI] [PubMed] [Google Scholar]
  • 94.Fruman DA.Phosphoinositide 3-kinase and its targets in B-cell and T-cell signaling. Curr Opin Immunol 2004; 16: 314–320. [DOI] [PubMed] [Google Scholar]
  • 95.Beşliu AN, Pistol G, Marica CM, et al. PI3K/Akt signaling in peripheral T lymphocytes from systemic lupus erythematosus patients. Roum Arch Microbiol Immunol 2009; 68: 69–79. [PubMed] [Google Scholar]
  • 96.Besliu A, Banica L, Matache C, et al. Systemic lupus erythematosus: the involvement of PI3K/Akt/mTOR pathway in cellular cycle and in early apoptosis. Joint Bone Spine 2008; 75: 246. [Google Scholar]
  • 97.Zhang Y, Zhao M, Sawalha AH, et al. Impaired DNA methylation and its mechanisms in CD4(+)T cells of systemic lupus erythematosus. J Autoimmun 2013; 41: 92–99. [DOI] [PubMed] [Google Scholar]
  • 98.Chen SY, Liu MF, Kuo PY, et al. Upregulated expression of STAT3/IL-17 in patients with systemic lupus erythematosus. Clin Rheumatol 2019; 38: 1361–1366. [DOI] [PubMed] [Google Scholar]
  • 99.Gao X, Liu L, Min X, et al. Non-coding RNAs in CD4+ T cells: new insights into the pathogenesis of systemic lupus erythematosus. Front Immunol 2020; 11: 568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang X, Ma R, Shi W, et al. Emerging roles of circular RNAs in systemic lupus erythematosus. Mol Ther Nuc Acid 2021; 24: 212–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Liu H, Zou Y, Chen C, et al. Current understanding of circular RNAs in systemic lupus erythematosus. Front Immunol 2021; 12: 628872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Fike AJ, Elcheva I, Rahman ZSM. The post-GWAS era: how to validate the contribution of gene variants in lupus. Curr Rheumatol Rep 2019; 21: 3. [DOI] [PubMed] [Google Scholar]
  • 103.Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75: 843–854. [DOI] [PubMed] [Google Scholar]
  • 104.Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993; 75: 855–862. [DOI] [PubMed] [Google Scholar]
  • 105.Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol 2019; 20: 21–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Qu B, Shen N.Chapter 27 – microRNA in systemic lupus erythematosus. In: Tsokos GC. (ed.) Systemic lupus erythematosus. Boston, MA: Academic Press, 2016, pp. 231–236. [Google Scholar]
  • 107.Zheng B, Zhang P, Yuan L, et al. Effects of human umbilical cord mesenchymal stem cells on inflammatory factors and miR-181a in T lymphocytes from patients with systemic lupus erythematosus. Lupus 2020; 29: 126–135. [DOI] [PubMed] [Google Scholar]
  • 108.Li HS, Ning Y, Li SB, et al. Expression and clinical significance of miR-181a and miR-203 in systemic lupus erythematosus patients. Eur Rev Med Pharmacol Sci 2017; 21: 4790–4796 [PubMed] [Google Scholar]
  • 109.He Y-L, Yang J, Zeng Z-N, et al. Interaction of miR-181b and IFNA1 polymorphisms on the risk of systemic lupus erythematosus. Biomed Res Int 2020; 2020: 4757065–4757065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Senousy MA, Helmy HS, Fathy N, et al. Association of MTMR3 rs12537 at miR-181a binding site with rheumatoid arthritis and systemic lupus erythematosus risk in Egyptian patients. Sci Rep 2019; 9: 12299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wang R, Wang CF, Qin HM, et al. Association between polymorphisms in the promoter region of miR-17-92 cluster and systemic lupus erythematosus in a Chinese population. J Cell Mol Med 2018; 22: 4016–4020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Motawi TK, Mohsen DA, El-Maraghy SA, et al. MicroRNA-21, microRNA-181a and microRNA-196a as potential biomarkers in adult Egyptian patients with systemic lupus erythematosus. Chem Biol Interact 2016; 260: 110–116. [DOI] [PubMed] [Google Scholar]
  • 113.Kourti M, Sokratous M, Katsiari CG.Regulation of microRNA in systemic lupus erythematosus: the role of miR-21 and miR-210. Mediterr J Rheumatol 2020; 31: 71–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Suo QF, Sheng J, Qiang FY, et al. 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: 345–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Xie L, Xu J.Role of MiR-98 and its underlying mechanisms in systemic lupus erythematosus. J Rheumatol 2018; 45: 1397–1405. [DOI] [PubMed] [Google Scholar]
  • 116.Wang G, Tam L-S, Kwan BC-H, et al. Expression of miR-146a and miR-155 in the urinary sediment of systemic lupus erythematosus. Clin Rheumatol 2012; 31: 435–440. [DOI] [PubMed] [Google Scholar]
  • 117.Shumnalieva R, Kachakova D, Shoumnalieva-Ivanova V, et al. Whole peripheral blood miR-146a and miR-155 expression levels in systemic lupus erythematosus patients. Acta Reumatol Port 2018; 43: 217–225. [PubMed] [Google Scholar]
  • 118.Zárate-Neira LÁ, Sánchez-Barbosa S, Pedroza-Torres A, et al. Dysregulation of miR-155-5p and miR-200-3p and the anti-non-bilayer phospholipid arrangement antibodies favor the development of lupus in three novel murine lupus models. J Immunol Res 2017; 2017: 8751642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.El-Shaer OS, Sabry JH, Mahgoub MY, et al. MiR-155 expression is a potential biomarker of systemic lupus erythematosus diagnosis and disease activity prediction. Meta Gene 2020; 26: 100770. [Google Scholar]
  • 120.Wang G, Tam LS, Li EK, et al. Serum and urinary cell-free MiR-146a and MiR-155 in patients with systemic lupus erythematosus. J Rheumatol 2010; 37: 2516–2522. [DOI] [PubMed] [Google Scholar]
  • 121.Stypinska B, Wajda A, Walczuk E, et al. The serum cell-free microRNA expression profile in MCTD, SLE, SSc, and RA patients. J Clin Med 2020; 9: 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Huang Q, Chen SS, Li J, et al. miR-210 expression in PBMCs from patients with systemic lupus erythematosus and rheumatoid arthritis. Ir J Med Sci 2018; 187: 243–249. [DOI] [PubMed] [Google Scholar]
  • 123.Nagy D, Shaheen NH, Selim HM, et al. MicroRNA-126 and 146a as potential biomarkers in systemic lupus erythematosus patients with secondary antiphospholipid syndrome. Egyptian Rheumatol 2020; 42: 201–206. [Google Scholar]
  • 124.Dominguez-Gutierrez PR, Ceribelli A, Satoh M, et al. Positive correlation of STAT1 and miR-146a with anemia in patients with systemic lupus erythematosus. J Clin Immunol 2014; 34: 171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Dong C, Zhou Q, Fu T, et al. Circulating exosomes derived-miR-146a from systemic lupus erythematosus patients regulates senescence of mesenchymal stem cells. Biomed Res Int 2019; 2019: 6071308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Labib DA, Koptan D, Ghoniem S, et al. Dysregulation of microRNA146a-5p expression in systemic lupus erythematosus females: diagnostic potential and association with ocular manifestations. Egypt Rheumatol 2020; 42: 117–121. [Google Scholar]
  • 127.Katsuyama E, Yan M, Watanabe KS, et al. Downregulation of miR-200a-3p, targeting CtBP2 complex, is involved in the hypoproduction of IL-2 in systemic lupus erythematosus-derived T cells. J Immunol 2017; 198: 4268–4276. [DOI] [PubMed] [Google Scholar]
  • 128.Cao W, Qian G, Luo W, et al. miR-125b is downregulated in systemic lupus erythematosus patients and inhibits autophagy by targeting UVRAG. Biomed Pharmacother 2018; 99: 791–797. [DOI] [PubMed] [Google Scholar]
  • 129.Wang X, Zhang C, Wu Z, et al. CircIBTK inhibits DNA demethylation and activation of AKT signaling pathway via miR-29b in peripheral blood mononuclear cells in systemic lupus erythematosus. Arthrit Res Ther 2018; 20: 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Shi X, Ye L, Xu S, et al. Downregulated miR-29a promotes B cell overactivation by upregulating Crk-like protein in systemic lupus erythematosus. Mol Med Rep 2020; 22: 841–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Su X, Ye L, Chen X, et al. MiR-199-3p promotes ERK-mediated IL-10 production by targeting poly (ADP-ribose) polymerase-1 in patients with systemic lupus erythematosus. Chem Biol Interact 2019; 306: 110–116. [DOI] [PubMed] [Google Scholar]
  • 132.Zhang Y, Chen X, Deng Y.miR-125a-3p decreases levels of interlukin-17 and suppresses renal fibrosis via down-regulating TGF-β1 in systemic lupus erythematosus mediated lupus nephritic mice. Am J Transl Res 2019; 11: 1843–1853. [PMC free article] [PubMed] [Google Scholar]
  • 133.Ramkissoon SH, Mainwaring LA, Ogasawara Y, et al. Hematopoietic-specific microRNA expression in human cells. Leuk Res 2006; 30: 643–647. [DOI] [PubMed] [Google Scholar]
  • 134.Carlsen AL, Schetter AJ, Nielsen CT, et al. Circulating microRNA expression profiles associated with systemic lupus erythematosus. Arthritis Rheum 2012; 65: 1324–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wang C-f, Wang R, Lei M, et al. Association of miR-181 cluster polymorphisms with systemic lupus erythematosus risk. Int J Clin Exp Med 2018; 11: 9388–9395. [Google Scholar]
  • 136.Johnson CD, Esquela-Kerscher A, Stefani G, et al. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res 2007; 67: 7713–7722. [DOI] [PubMed] [Google Scholar]
  • 137.Fu Y, Liu X, Chen Q, et al. Downregulated miR-98-5p promotes PDAC proliferation and metastasis by reversely regulating MAP4K4. J Exp Clin Cancer Res 2018; 37: 130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Hu C, Huang S, Wu F, et al. miR-98 inhibits cell proliferation and induces cell apoptosis by targeting MAPK6 in HUVECs. Exp Ther Med 2018; 15: 2755–2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Jiang F, Yu Q, Chu Y, et al. MicroRNA-98-5p inhibits proliferation and metastasis in non-small cell lung cancer by targeting TGFBR1. Int J Oncol 2019; 54: 128–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Shi XY, Wang H, Wang W, et al. MiR-98-5p regulates proliferation and metastasis of MCF-7 breast cancer cells by targeting Gab2. Eur Rev Med Pharmacol Sci 2020; 24: 10914. [DOI] [PubMed] [Google Scholar]
  • 141.Wang Z, Han Z, Zhang L, et al. MicroRNA-98-5p regulates the proliferation and apoptosis of A549 cells by targeting MAP4K3. Oncol Lett 2019; 18: 4288–4293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ji ML, Lu J, Shi PL, et al. Dysregulated miR-98 contributes to extracellular matrix degradation by targeting IL-6/STAT3 signaling pathway in human intervertebral disc degeneration. J Bone Miner Res 2016; 31: 900–909. [DOI] [PubMed] [Google Scholar]
  • 143.Yuan S, Tang C, Chen D, et al. miR-98 modulates cytokine production from human PBMCs in systemic lupus erythematosus by targeting IL-6 mRNA. J Immunol Res 2019; 2019: 9827574–9827574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Tam W.Identification and characterization of human BIC, a gene on chromosome 21 that encodes a noncoding RNA. Gene 2001; 274: 157–167. [DOI] [PubMed] [Google Scholar]
  • 145.Kim VN, Han J, Siomi MC.Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol 2009; 10: 126–139. [DOI] [PubMed] [Google Scholar]
  • 146.Leng RX, Pan HF, Qin WZ, et al. Role of microRNA-155 in autoimmunity. Cytokine Growth Factor Rev 2011; 22: 141–147. [DOI] [PubMed] [Google Scholar]
  • 147.Leiss H, Salzberger W, Jacobs B, et al. MicroRNA 155-deficiency leads to decreased autoantibody levels and reduced severity of nephritis and pneumonitis in pristane-induced lupus. PLoS ONE 2017; 12: e0181015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Latini A, Ciccacci C, Benedittis G, 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] [PubMed] [Google Scholar]
  • 149.Xu WD, Lu MM, Pan HF, et al. Association of MicroRNA-146a with autoimmune diseases. Inflammation 2012; 35: 1525–1529. [DOI] [PubMed] [Google Scholar]
  • 150.Fu HX, Fan XP, Li M, et al. MiR-146a relieves kidney injury in mice with systemic lupus erythematosus through regulating NF-κB pathway. Eur Rev Med Pharmacol Sci 2019; 23: 7024–7032. [DOI] [PubMed] [Google Scholar]
  • 151.Zundel W, Schindler C, Haas-Kogan D, et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev 2000; 14: 391–396. [PMC free article] [PubMed] [Google Scholar]
  • 152.Devlin C, Greco S, Martelli F, et al. miR-210: more than a silent player in hypoxia. IUBMB Life 2011; 63: 94–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Huang X, Le QT, Giaccia AJ.MiR-210 – micromanager of the hypoxia pathway. Trends Mol Med 2010; 16: 230–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhang Z, Sun H, Dai H, et al. MicroRNA miR-210 modulates cellular response to hypoxia through the MYC antagonist MNT. Cell Cycle 2009; 8: 2756–2768. [DOI] [PubMed] [Google Scholar]
  • 155.Garchow B, Maque Acosta Y, Kiriakidou M.HIF-1α and miR-210 differential and lineage-specific expression in systemic lupus erythematosus. Mol Immunol 2021; 133: 128–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wang S, Wan X, Ruan Q.The microRNA-21 in autoimmune diseases. Int J Mol Sci 2016; 17: 864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Sheedy FJ, Palsson-McDermott E, Hennessy EJ, et al. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat Immunol 2010; 11: 141–147. [DOI] [PubMed] [Google Scholar]
  • 158.Wang L, He L, Zhang R, et al. Regulation of T lymphocyte activation by microRNA-21. Mol Immunol 2014; 59: 163–171. [DOI] [PubMed] [Google Scholar]
  • 159.Sheedy FJ.Turning 21: induction of miR-21 as a key switch in the inflammatory response. Front Immunol 2015; 6: 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. https://www.ncbi.nlm.nih.gov/gene/407021
  • 161.Di Marco M, Ramassone A, Pagotto S, et al. MicroRNAs in autoimmunity and hematological malignancies. Int J Mol Sci 2018; 19: 3139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zeng HF, Yan S, Wu SF.MicroRNA-153-3p suppress cell proliferation and invasion by targeting SNAI1 in melanoma. Biochem Biophys Res Commun 2017; 487: 140–145. [DOI] [PubMed] [Google Scholar]
  • 163.Li D, Li X, Duan M, et al. MiR-153-3p induces immune dysregulation by inhibiting PELI1 expression in umbilical cord-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Autoimmunity 2020; 53: 201–209. [DOI] [PubMed] [Google Scholar]
  • 164.Wang Y, Zeng G, Jiang Y.The emerging roles of miR-125b in cancers. Cancer Manag Res 2020; 12: 1079–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Ren K, Li T, Zhang W, et al. miR-199a-3p inhibits cell proliferation and induces apoptosis by targeting YAP1, suppressing Jagged1-Notch signaling in human hepatocellular carcinoma. J Biomed Sci 2016; 23: 79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Pang B, Zhen Y, Hu C, et al. Myeloid-derived suppressor cells shift Th17/Treg ratio and promote systemic lupus erythematosus progression through arginase-1/miR-322-5p/TGF-β pathway. Clin Sci 2020; 134: 2209–2222. [DOI] [PubMed] [Google Scholar]
  • 167.Cheng T, Ding S, Liu S, et al. Resolvin D1 improves the Treg/Th17 imbalance in systemic lupus erythematosus through miR-30e-5p. Front Immunol 2021; 12: 668760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Gao Y, Li S, Zhang Z, et al. The role of long non-coding RNAs in the pathogenesis of RA, SLE, and SS. Front Med 2018; 5: 193–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Bierhoff H, Schmitz K, Maass F, et al. Noncoding transcripts in sense and antisense orientation regulate the epigenetic state of ribosomal RNA genes. Cold Spring Harb Symp Quant Biol 2010; 75: 357–364. [DOI] [PubMed] [Google Scholar]
  • 170.Peng WX, Koirala P, Mo YY.LncRNA-mediated regulation of cell signaling in cancer. Oncogene 2017; 36: 5661–5667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Wang C, Wang L, Ding Y, et al. LncRNA structural characteristics in epigenetic regulation. Int J Mol Sci 2017; 18: 2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Marques-Rocha JL, Samblas M, Milagro FI, et al. Noncoding RNAs, cytokines, and inflammation-related diseases. FASEB J 2015; 29: 3595–3611. [DOI] [PubMed] [Google Scholar]
  • 173.Maass PG, Luft FC, Bähring S.Long non-coding RNA in health and disease. J Mol Med 2014; 92: 337–346. [DOI] [PubMed] [Google Scholar]
  • 174.Zhang Q, Liang Y, Yuan H, et al. Integrated analysis of lncRNA, miRNA and mRNA expression profiling in patients with systemic lupus erythematosus. Arch Med Sci 2019; 15: 872–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ye H, Wang X, Wang L, et al. Full high-throughput sequencing analysis of differences in expression profiles of long noncoding RNAs and their mechanisms of action in systemic lupus erythematosus. Arthritis Res Ther 2019; 21: 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Li LJ, Zhao W, Tao SS, et al. Comprehensive long non-coding RNA expression profiling reveals their potential roles in systemic lupus erythematosus. Cell Immunol 2017; 319: 17–27. [DOI] [PubMed] [Google Scholar]
  • 177.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: 646–652. [PMC free article] [PubMed] [Google Scholar]
  • 178.Zhang F, Wu L, Qian J, et al. Identification of the long noncoding RNA NEAT1 as a novel inflammatory regulator acting through MAPK pathway in human lupus. J Autoimmun 2016; 75: 96–104. [DOI] [PubMed] [Google Scholar]
  • 179.Cao H-Y, Li D, Wang Y-P, et al. Clinical significance of reduced expression of lncRNA TUG1 in the peripheral blood of systemic lupus erythematosus patients. Int J Rheum Dis 2020; 23: 428–434. [DOI] [PubMed] [Google Scholar]
  • 180.Cao HY, Li D, Wang YP, et al. The protection of NF-κB inhibition on kidney injury of systemic lupus erythematosus mice may be correlated with lncRNA TUG1. Kaohsiung J Med Sci 2020; 36: 354–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Liu CH, Lu YL, Huang HT, 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: 3548–3559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Liu Q, Deng Y, Li C, 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: 41–47. [DOI] [PubMed] [Google Scholar]
  • 183.Wu G-C, Li J, Leng R-X, 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: 23650–23663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Li J, Wu G-C, Zhang T-P, et al. Association of long noncoding RNAs expression levels and their gene polymorphisms with systemic lupus erythematosus. Sci Rep 2017; 7: 15119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Wang JB, Li J, Zhang TP, et al. Expression of several long noncoding RNAs in peripheral blood mononuclear cells of patients with systemic lupus erythematosus. Adv Med Sci 2019; 64: 430–436. [DOI] [PubMed] [Google Scholar]
  • 186.Yang H, Liang N, Wang M, et al. Long noncoding RNA MALAT-1 is a novel inflammatory regulator in human systemic lupus erythematosus. Oncotarget 2017; 8: 77400–77406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Taraschi TF, Ellingson JS, Rubin E.Membrane structural alterations caused by chronic ethanol consumption: the molecular basis of membrane tolerance. Ann N Y Acad Sci 1987; 492: 171–180. [DOI] [PubMed] [Google Scholar]
  • 188.Dong P, Xiong Y, Yue J, et al. Long non-coding RNA NEAT1: a novel target for diagnosis and therapy in human tumors. Front Genet 2018; 9: 471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Chen L, Niu Q, Huang Z, et al. IKZF1 polymorphisms are associated with susceptibility, cytokine levels, and clinical features in systemic lupus erythematosus. Medicine 2020; 99: e22607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Dong G, Yang Y, Li X, 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] [PubMed] [Google Scholar]
  • 191.Li Z, Shen J, Chan MT, et al. TUG1: a pivotal oncogenic long non-coding RNA of human cancers. Cell Prolif 2016; 49: 471–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Xu Y, Deng W, Zhang W.Long non-coding RNA TUG1 protects renal tubular epithelial cells against injury induced by lipopolysaccharide via regulating microRNA-223. Biomed Pharm 2018; 104: 509–519. [DOI] [PubMed] [Google Scholar]
  • 193.Zhang H, Li H, Ge A, et al. Long non-coding RNA TUG1 inhibits apoptosis and inflammatory response in LPS-treated H9c2 cells by down-regulation of miR-29b. Biomed Pharm 2018; 101: 663–669. [DOI] [PubMed] [Google Scholar]
  • 194. https://www.ncbi.nlm.nih.gov/gene/60674
  • 195.Wu GC, Hu Y, Guan SY, et al. Differential plasma expression profiles of long non-coding RNAs reveal potential biomarkers for systemic lupus erythematosus. Biomolecules 2019; 9: 206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Wang P, Xue Y, Han Y, et al. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 2014; 344: 310–313. [DOI] [PubMed] [Google Scholar]
  • 197.Chen Y, Chen Y, Zu B, et al. Identification of long noncoding RNAs lnc-DC in plasma as a new biomarker for primary Sjögren’s syndrome. J Immunol Res 2020; 2020: 9236234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Wang J, Wei F, Zhou H.Advances of lncRNA in autoimmune diseases. Front Lab Med 2018; 2: 79–82. [Google Scholar]
  • 199.Da C-m, Gong C-Y, Nan W, et al. The role of long non-coding RNA MIAT in cancers. Biomed Pharmacother 2020; 129: 110359. [DOI] [PubMed] [Google Scholar]
  • 200.Xu H, Chen W, Zheng F, et al. Reconstruction and analysis of the aberrant lncRNA-miRNA-mRNA network in systemic lupus erythematosus. Lupus 2020; 29: 398–406. [DOI] [PubMed] [Google Scholar]
  • 201.Zhang Y, Xie L, Lu W, 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] [PMC free article] [PubMed] [Google Scholar]
  • 202.https://www.genecards.org/cgi-bin/carddisp.pl?gene=MALAT1
  • 203.Amodio N, Raimondi L, Juli G, et al. MALAT1: a druggable long non-coding RNA for targeted anti-cancer approaches. J Hematol Oncol 2018; 11: 63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Kristensen LS, Andersen MS, Stagsted LVW, et al. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet 2019; 20: 675–691. [DOI] [PubMed] [Google Scholar]
  • 205.Sanger HL, Klotz G, Riesner D, et al. Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA 1976; 73: 3852–3856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Li J, Sun D, Pu W, et al. Circular RNAs in cancer: biogenesis, function, and clinical significance. Trends Cancer 2020; 6: 319–336. [DOI] [PubMed] [Google Scholar]
  • 207.Shang Q, Yang Z, Jia R, et al. The novel roles of circRNAs in human cancer. Mol Cancer 2019; 18: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Wang M, Yu F, Li P, et al. Emerging function and clinical significance of exosomal circRNAs in cancer. Mol Ther: Nucl Acid 2020; 21: 367–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Cheng J, Meng J, Zhu L, et al. Exosomal noncoding RNAs in glioma: biological functions and potential clinical applications. Mol Cancer 2020; 19: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Shi X, Wang B, Feng X, et al. circRNAs and exosomes: a mysterious frontier for human cancer. Mol Ther: Nucl Acid 2020; 19: 384–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Liu J, To AC.Quantitative texture prediction of epitaxial columnar grains in additive manufacturing using selective laser melting. Addit Manuf 2017; 16: 58–64. [Google Scholar]
  • 212.Cortes R, Forner MJ.Circular RNAS: novel biomarkers of disease activity in systemic lupus erythematosus? Clin Sci 2019; 133: 1049–1052. [DOI] [PubMed] [Google Scholar]
  • 213.Luo Q, Zhang L, Xiong L, 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: 822–833. [PubMed] [Google Scholar]
  • 214.Lin S, Fan R, Li W, et al. A ceRNA regulatory network in systemic lupus erythematosus and its molecular interplay with cancer. Ann Transl Med 2022; 10: 563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Zhang J, Liu Y, Shi G.The circRNA-miRNA-mRNA regulatory network in systemic lupus erythematosus. Clin Rheumatol 2021; 40: 331–339. [DOI] [PubMed] [Google Scholar]
  • 216.Zheng F, Yu X, Tang D, et al. The identification of circular RNAs from peripheral blood mononuclear cells in systemic lupus erythematosus. BMC Med Genomics 2021; 14: 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Miao Q, Zhong Z, Jiang Z, et al. RNA-seq of circular RNAs identified circPTPN22 as a potential new activity indicator in systemic lupus erythematosus. Lupus 2019; 28: 520–528. [DOI] [PubMed] [Google Scholar]
  • 218.Li LJ, Zhu ZW, Zhao W, et al. Circular RNA expression profile and potential function of hsa_circ_0045272 in systemic lupus erythematosus. Immunology 2018; 155: 137–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Zhang MY, Wang JB, Zhu ZW, et al. Differentially expressed circular RNAs in systemic lupus erythematosus and their clinical significance. Biomed Pharmacother 2018; 107: 1720–1727. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Chronic Disease are provided here courtesy of SAGE Publications

RESOURCES