Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2024 May 29.
Published in final edited form as: Curr Opin Rheumatol. 2024 Feb 29;36(3):191–200. doi: 10.1097/BOR.0000000000001008

Monogenic Lupus: Insights into Disease Pathogenesis and Therapeutic Opportunities

Yuting Qin 1,#, Jianyang Ma 1,#, Carola G Vinuesa 1,2,
PMCID: PMC7616038  EMSID: EMS196092  PMID: 38420886

Abstract

Purpose of review

This review aims to provide an overview of the genes and molecular pathways involved in monogenic lupus, the implications for genome diagnosis, and the potential therapies targeting these molecular mechanisms.

Recent findings

To date, more than 30 genes have been identified as contributors to monogenic lupus. These genes are primarily related to complement deficiency, activation of the type I interferon (IFN) pathway, disruption of B cell and T cell tolerance and metabolic pathways, which reveal the multifaceted nature of systemic lupus erythematosus (SLE) pathogenesis.

Summary

In–depth study of the causes of monogenic lupus can provide valuable insights into of pathogenic mechanisms of SLE, facilitate the identification of effective biomarkers, and aid in developing therapeutic strategies.

Keywords: Systemic lupus erythematosus, monogenic lupus, complement deficiency, type I interferon, immunotolerance

Introduction

SLE is a systemic autoimmune disease that predominantly affects women, impacting multiple organs such as the kidneys, skin, joints, lungs, cardiovascular system, central nervous system, and hematopoietic system (1). Although the etiology of SLE remains to be fully elucidated, there is increasing realization that it is a disorder of increased nucleic acid sensing or signaling, with genetic and environmental factors contributing to its onset (1). A significant genetic component is underscored by twin studies showing higher concordance rates in monozygotic twins compared to dizygotic twins and familial aggregation (2, 3).

SLE is primarily a polygenic disease, with numerous risk variants identified through genome-wide association studies (GWAS) and an increasing number of functional rare variants identified through whole exome sequencing (WES) and whole genome sequencing (WGS) studies (4). Variants identified by GWAS are common in the general population, and although most have small or modest effects, they can act additively to increase the risk of developing SLE (5). However, risk alleles identified by GWAS still account for only a small fraction of the genetic risk associated with disease (6). It is increasingly acknowledged that additional more rare variants may contribute to the missing heritability and exert strong effects. These rare variants can occur in GWAS SLE risk genes, or in genes known to cause monogenic lupus, or in genes not yet associated with SLE (7). In recent years, the increasing accessibility and affordability of next-generation sequencing technologies such as WES and WGS have enabled the discovery of novel and rare variants, expanding the list of genes causing monogenic SLE (4). Monogenic lupus, associated with alterations in single genes, is a rare yet severe form of SLE that typically manifests during childhood (8). Despite constituting a small fraction of lupus cases, monogenic lupus greatly enhances our understanding of disease pathogenesis and illuminates therapeutic targets. This article aims to review our current knowledge of the genes involved in monogenic lupus and their corresponding molecular pathways. It outlines a growing role of genomic diagnosis and highlights potential targeted therapies derived from understanding the molecular mechanisms.

Genetic basis of monogenic lupus

The pathogenesis of SLE is complex, encompassing abnormalities across all facets of the immune system. Central to SLE pathogenesis is a breakdown in B cell tolerance to nucleic acids and activation of the type I IFN pathway and complement. Most gene variants implicated in monogenic lupus relate to these interconnected processes. (Table 1 & Figure 1).

Table 1. Genes causing monogenic lupus.

Pathway Gene (protein)a MIM Associated disease Number of reports of monogenic lupus (published year) (ref) b
Complement activation C1QA/C1QB/C1/QC C1Q deficiency >30
C1R/C1S Ehlers-Danlos syndrome type 1;C1S deficiency 7
C2 C2 deficiency >20
C4A/C4B C4a and C4b deficiencies >20
C3 C3 deficiency 7
FCN3 Immunodeficiency due to ficolin3 deficiency 2 (in 2019, 2020) (12, 13)
Activation of type 1 IFN pathway DNASE1 SLE 1 (in 2001) (28)
DNASE1L3 SLE and HUV 10
DNASE2 Autoinflammatory-pancytopenia syndrome 3 (in 2020, 2021, 2023) (2931)
TREX1 AGS, FCL, SSd, SS 16
RNASEH2A/B/C (RNASEH2) AGS 1 (in 2015) (37)
SAMHD1 AGS, FCL, CLL (somatic) 5 (in 2011, 2018, 2021, 2022) (3236)
CECR1 (ADA2) DADA2 2 (in 2016, 2017) (42, 43)
SAT1 SLE 1 (in 2022) (44)
DDX58 (RIG–I) SMS 1 (in 2023) (49)
TMEM173 (STING) SAVI, FCL 2 (in 2014, 2017) (47, 48)
IFIH1 (MDA5) AGS, FCL, SMS 1 (in 2015) (46)
TLR7 SLE 1 (in 2022) (45)
UNC93B1 HSE 2 (in 2024) (52,53)
IKZF1 (IKAROS) CVID, ITP 2 (in 2017, 2019) (61, 62)
TNFAIP3 (A20) Autoinflammatory syndrome, Behçet-like 3 (in 2019, 2020, 2021) (6567)
ACP5 (TRAP) SPENCD 8
PACSIN1 NA 1 (in 2023) (56)
NLRC4 Familial cold autoinflammatory syndrome, Autoinflammation with infantile enterocolitis 1 (in 2023) (58)
ISG15 Immunodeficiency 1 (in 2021) (70)
T cell and B cell tolerance FAS/FASLG ALPS 1 (in 1996) (71)
PRKCD (PKC delta) ALPS 5 (in 2013, 2015, 2017, 2018, 2022) (7276)
PTPN11/RAF/K RAS/NRAS/SHO C2 Noonan syndrome 9
PIK3CD (PI3K p110 δ) Immunodeficiency 5 (in 2019, 2020, 2021, 2023) (29, 66, 7981)
DOCK8 Hyper–IgE syndrome, autosomal recessive, with recurrent infections 3 (in 2014, 2021) (8587)
SH2B3 Erythrocytosis, Myelofibrosis, Thrombocythemia 1 (in 2024)(86)
P2RY8 NA 1 (in 2022) (83)
BACH2 Immunodeficiency and autoimmunity 1 (in 2023) (90, 91)
LRBA Immunodeficiency, common variable, with autoimmunity 1 (in 2020) (88)
metabolism CYBB (NOX2) CGD 7
SLC7A7 LPI 6
PEPD Prolidase deficiency 7
MAN2B1 α-Mannosidosis, types I and II 3 (in 2004, 2019, 2022) (9597)

HUV, hypocomplementaemic urticarial vasculitis; SSd, Sjögren syndrome; SS, systemic sclerosis; CLL, chronic lymphocytic leukaemia; HSE, herpes simplex encephalitis; ITP, immune thrombocytopenic purpura; NA, not available.

a

Protein name specified in instances where it differs from the gene name.

b

If the number of reports on gene variants leading to monogenic lupus is less than or equal to 5, the published year and references will be listed.

Figure 1.

Figure 1

Signaling pathways involved in the pathogenesis of monogenic lupus. (Created with BioRender.com)

Complement activation, nucleic acid degradation, nucleic acid sensing and signaling, as well as type I IFN signaling, are the main molecular pathways involved in the development of monogenic lupus. LoF variants in genes encoding proteins related to complement activation (including C1Q, C1R/C1S, C2, C4, and FCN3) and extracellular nucleic acid degradation (DNASE1, DNASE1L3, and SAT1) prevent clearance of apoptotic cellular debris, mitochondrial DNA, and neutrophil extracellular traps. Accumulation of nucleic acids stimulates the type I IFN pathway. This pathway can also be overactivated by LoF variants in genes encoding proteins linked to intracellular nucleic acid sensing and/or degradation (including DNASE2, TREX1, SAMHD1, RNASEH2, c-GAS/STING, RIG-I, and MDA5). The endosomal ssRNA receptor TLR7 can also lead to SLE, and TLR7 signaling can be restrained by TLR9. Variants in genes that regulate TLR7/MyD88 and/or type I IFN signaling (such as ACP5, PACSIN1, IKAROS, A20, ISG15, UNC93B1 and NLRC4) have also been shown to cause SLE. Proteins in red indicate GoF (Gain of Function); green indicate LoF (Loss of Function). Dashed lines: mechanism is still uncertain.

Complement deficiency

The complement system, consisting of a complex network of proteins and receptors, is crucial for host defense against pathogens and maintenance of homeostasis by removing immune complexes and cell debris. It can be activated through various triggers and operates mainly via three pathways: the classical, lectin, and alternative pathway. Each pathway concludes with the creation of C3 convertases and shares a common final step leading to the assembly of the membrane-attack complex (MAC), which creates a pore in the target cell, promoting osmotic cell lysis.

Homozygous deficiencies or loss-of-function (LoF) variants in the early components of the classical pathway were the first identified causes of monogenic SLE (9). This pathway begins with C1q binding to immune complexes, sequentially activating C1R and C1S to form the C1 complex. Once this complex is formed, C1S activates C4 and C2, resulting in the formation of the C3 convertase (also known as C4b2a) (10). The incidence rate of SLE or SLE-like phenotypes in the presence of C1Q, C1R/C1S, C4, and C2 deficiencies is approximately 90%, 65%, 75%, and 10% respectively (11). In addition, another complement protein in the lectin pathway, FICOLIN-3 (encoded by FCN3), has also been linked to SLE. FCN3 deficiency can increase the risk of developing SLE, with an incidence rate of around 30% (12, 13).

At least three mechanisms have been proposed to explain how deficiency in complement components causes SLE. The first one relates to ineffective clearance of apoptotic debris, which exposes nucleic acid autoantigens and increases their burden. Indeed, C1Q, C1R, C1S, C2, C3 and C4 can mediate opsonization of apoptotic cells. C1Q can also directly bind to apoptotic cells and ligate calreticulin and CD91 on phagocytes, thus enabling phagocytosis (14). A second mechanism involves negative regulation of type I IFN production by C1Q. This is achieved by C1Q preferentially promoting the binding of immune complexes to monocytes and thus preventing these complexes from attaching to, and activating, plasmacytoid dendritic cells (pDCs), which are a major source of type I IFN (15). The third mechanism involves the role of complement receptors 1 (CR1/CD35) and 2 (CR2/CD21) in maintaining immune tolerance through B cell intrinsic or extrinsic mechanisms. In the hen egg lysozyme (HEL) model, CR1/CR2 or C4 deficient self-reactive B cells fail to undergo anergy (16). One possible explanation is that CD21 can form a co–receptor with CD19 and CD81 on B cells to lower the threshold to BCR-signaling (17). CR1/CR2 are first expressed at the T1-T2 transitional stage in which a lower threshold of BCR signal can increase the sensitivity of the transitional B cell to C4b coated self-antigens and result in cell death (18). Apart from the expression on B cells, CR1/CR2 are highly expressed on follicular dendritic cells (FDCs), and help these cells to bind and present C3d-opsonized auto-antigen, thereby delivering ligands to autoreactive B cells circulating through follicles, which die shortly afterwards in the absence of T cell help (19). More recent studies have suggested additional protective roles of complement in SLE. For instance, C1q has been found to modulate the mitochondrial metabolism of CD8(+) T cells, thus inhibiting autoreactive responses to self-antigens (20).

Despite the overall protective effects of complement with regards to SLE development, detrimental roles of C1Q in SLE have also been suggested: C1Q appears to prevent the degradation of neutrophil extracellular traps (NETs), which can induce type I IFN (21), and mediates microglia-driven destruction of synaptic terminals in neuropsychiatric lupus (22).

Activation of the type I IFN pathway

Type I IFN, including IFN–α and IFN–β, play a crucial role in the protective immune response against viral infections and the regulation of immune cell functions. Activation of type I IFN signaling is a hallmark of SLE (23). Moreover, several monogenic forms of lupus are classified as type I interferonopathies due to their prominent type I IFN signature (24). There are three distinct mechanisms through which the overactivation of type I IFN pathway can occur:

Defects in nucleic acid metabolism

Nucleic acid metabolism plays crucial roles in various biological processes, and includes DNA and RNA degradation, DNA replication, DNA repair, and RNA editing. Defects in nucleic acid metabolism lead to the accumulation of self-nucleic acids, which can initiate autoimmunity.

Lupus–relevant nucleic acids can derive from both extracellular and intracellular sources. Extracellular DNA usually generates through processes such as apoptosis, efferocytosis, NETosis, or extrusion of mitochondrial DNA from neutrophils, eosinophils and platelets (23). DNASE1L3 digests double-stranded DNA (dsDNA) derived from chromatin within apoptotic cell microparticles (25). Loss-of-function variants of this enzyme has been reported to cause SLE (26). DNASE1 degrades dsDNA present in NETs (27) but its role in SLE is less clear with a single case reported in 2001 (28).

Intracellular nucleic acids originate from replication stress, DNA or mitochondrial damage, and endogenous retroelement activation (23). Variants in genes involved in DNA or dNTP degradation (DNASE2(2931), TREX1 and SAMHD1(3236)) and RNA degradation (RNASEH2A, RNASEH2B and RNASEH2C(37)) (reviewed in 2018 (38)), are often associated with elevated levels of IFN–α and the emergence of familial chilblain lupus (FCL) (39) and childhood-onset SLE, as well as the classical interferonopathy Aicardi-Goutières syndrome (AGS) (24). Notably, CECR1, encoding adenosine deaminase 2 (ADA2), which when defective causes deficiency of adenosine deaminase 2 (DADA2) and functions as an extracellular deaminase (40), has been recently described as a lysosomal DNase and an activator of type I IFNs (41). Interestingly, two reports show several DADA2 patients exhibit manifestations like SLE (42, 43), with one report identifying abnormal activation of type I IFN in two sibling patients (42). Another protein that when deficient causes monogenic SLE is SAT1 (44). SAT1 encodes a rate-limiting enzyme in polyamine catabolism. Excessive polyamines may trigger SLE through their ability to bind DNA and induce NETosis; SAT1 is highly expressed in neutrophils (44).

Increased sensitivity, constitutive activation, or increased signaling from nucleic acids sensors

Extracellular and intracellular nucleic acids can be sensed by a set of pattern recognition receptors (PRR) including endosomal TLR7 and TLR9, as well as cytoplasmic STING (encoded by TMEM173), RIG-I (encoded by DDX58), and MDA5 (encoded by IFIH1). Gain-of-function (GoF) variants in TLR7 (45) and IFIH1 (46) have been reported to cause monogenic SLE as a result of enhanced sensitivity or constitutive activation, leading to a prominent type I IFN signature. TLR7 GoF has been shown to cause a B cell–intrinsic breach in tolerance (45) promoting the survival of BCR-activated cells and differentiation into CD11c+ age-associated B cells (ABCs) and plasma cells via an extrafollicular pathway. Additionally, there have been two reports with GoF variants in TMEM173 causing inflammatory syndrome and lupus-like disease (47), as well as FCL (48). Furthermore, a study identified a novel DDX58 GoF variant in five unrelated families associated with lupus nephritis (49). However, it should be noted that most variants in TMEM173 (50) and DDX58 (51) have been reported to cause STING-associated vasculopathy with onset in infancy (SAVI) and Singleton-Merten syndrome (SMS), respectively.

More recently, LoF variants in UNC93B1, which is important for endosomal transport and degradation of TLR7, has been reported to cause monogenic lupus (52, 53). Additional causes or contributors to monogenic SLE that are potentially linked with increased nucleic acid sensing include LoF variants in ACP5 and PACSIN1, and GoF variants in NLRC4. ACP5 encodes for tartrate-resistant acid phosphatase (TRAP), which controls the phosphorylation of osteopontin (OPN), a cytokine necessary to produce type I IFN following TLR7/9 and RIG-I like receptors (RLRs) stimulation (54, 55). LoF variants in ACP5 have been associated with Spondyloenchondrodysplasia (SPENCD), a rare skeletal disorder that often presents with lupus-like characteristics (56). PACSIN1 limits TLR7 signaling through disrupting TRAF4-mediated inhibition of TRAF6 (57). NLRC4, a member of the NOD-like receptor family that regulates inflammasome activation has also been reported to be a positive regulator of TBK1, thereby increasing IFN-β production (58), and causing SLE when overactive (59).

Other negative regulators of TLR/MyD88 signaling have also been shown to be critical for survival of self-reactive B cells and cause SLE when defective. These include IKZF1 and TNFAIP3. IKZF1, encoding the crucial hematopoietic transcription factor IKAROS, has also been linked to SLE, through defective transcription of negative regulators of MyD88 (60, 61). Although IKZF1 mutations are found in common variable immunodeficiency (CVID), there have been at least two reports of patients with IKZF1 LoF variants developing SLE (62, 63). TNFAIP3 encodes the A20 protein, a ubiquitin modifying enzyme that negatively regulates TLR7 signals leading to NF-kB activation (64). A20’s role in limiting B cell survival appears crucial to prevent autoimmunity (65). Three reports of patients with lupus described TNFAIP3 variants impairing A20-mediated deubiquitination, thought to be causal (6668).

Aberrant type I IFN signaling transduction

Nucleic acid sensors trigger a signaling cascade upon activation involving MYD88/TRAF6 or TBK1 and either IRF7, IRF5, or IRF3, leading to the production of type I IFNs. These interferons bind to receptors IFNAR1 and IFNAR2, subsequently activating the Janus kinase (JAK)-signal transducer and activator of transcription (STAT1/2) signaling pathway, which initiates the expression of IFN-stimulated genes (ISGs) (69). Notably, TLR7 and STAT1/2 are also ISGs, establishing a positive feedback loop that enhances nucleic acid sensing and antigen-presenting capacity, as well as supporting plasma cell differentiation and autoantibody production (23).

Several genes encoding proteins involved in the regulation of type I IFN signaling have been linked to SLE. For example, LoF variants in ISG15, which controls the stability of USP18 (70) – a negative regulator for IFN signaling, often cause AGS. However, there is a case report of ISG15 deficiency causing monogenic lupus (71).

Breakdown of B and T cell tolerance

The hallmark of SLE is a loss of B cell tolerance, resulting in the inability to eliminate autoreactive B cells, which go on to differentiate into autoantibody-secreting plasma cells. Loss of T cell tolerance and increased nucleic acid signals can also cause or contribute to a breach in B cell tolerance, as exemplified above with the TLR7 GoF variants.

Monogenic conditions that typically cause autoimmune lymphoproliferative syndrome (ALPS), may present with SLE-like disease (7277). ALPS can be caused by germline or somatic LoF variants in the FAS and FASLG genes, and LoF variants in PRKCD. FAS and FASL mediate activation-induced cell death, and their deficiency prevents elimination of chronically activated – including autoreactive – B and T cells (72). ALPS patients often present with enlarged lymph nodes, spleen, and liver and a different spectrum of autoantibodies to that found in SLE (78). PRKCD deficiency is characterized by enhanced B cell proliferation and survival and impaired negative selection of self-reactive B cells (77).

RASopathies, like Noonan syndrome, are another group of diseases linked to lymphocyte apoptosis dysregulation. Clinical manifestations include distinctive facial features, short stature, heart issues, developmental delays, and occasionally, SLE-like disease (79). These conditions can be caused by variants in genes encoding RAS/MAPK pathway signaling proteins, including PTPN11, RAF, KRAS, NRAS, and SHOC2.

In addition, variants in genes that regulate lymphocyte development, activation, selection, and migration can also cause complex syndromes presenting with both immunodeficiency and autoimmunity that may present with lupus-like manifestations. GoF mutations in PIK3CD (29, 67, 8082), that signals via AKT and mTOR to control the development and activation of T and B cells, result in a complex immunodeficiency syndrome referred to as activated PI3K δ syndrome, characterized by recurrent infections, lymphoproliferation, autoimmunity, and an increased risk of lymphoid malignancies (83). LoF variants of P2RY8 associated with increased AKT and ERK activity, have also been found to impair B cell tolerance and found to be likely causes or contributors to SLE (84). B cells carrying pathogenic P2RY8 variants could not be confined in germinal centers and negative selection of autoreactive B cells was impaired. DOCK8, a guanine nucleotide exchange factor predominantly expressed in lymphocytes, influences the actin cytoskeleton and lymphocyte migration and the activation of STAT3 (85). Hypomorphic variants in SH2B3 that break B cell tolerance at least in part through increased IFN-γ and IL-4 signaling have also been recently reported to contribute to lupus-pathogenesis in humans and mice (86). Deficiency in DOCK8 also leads to a combined immunodeficiency characterized by various features, including recurrent infections, autoimmunity and cancer (85), was associated with SLE in three case reports (8789). Another case report (90) has also described a variation in LRBA, which regulates CTLA4 intracellular trafficking and protein expression (91) that was associated with a complex childhood-onset autoimmunity including colitis, type 1 diabetes and SLE. BACH2 is a crucial transcription gene regulating T and B cell development, and its deficiency has been linked to SLE in mouse studies (92). Recently, a case of early-onset SLE in a BRIDA (BACH2-related immunodeficiency and autoimmunity) patient has suggested that BACH2 variants may constitute a potential monogenic cause of SLE (93).

Metabolic defects

Defects in several metabolic pathways have sporadically been associated with monogenic lupus-like phenotypes. For example, LoF variants in CYBB, which encodes NOX2, a component of NADPH oxidase, typically cause chronic granulomatous disease (CGD) and a proportion of patients develop SLE (94). Defects in SLC7A7 (encoding a subunit of a cationic amino acid transporter) can lead to lysinuric protein intolerance (LPI) and SLE (95). Deficiency in PEPD (encoding peptidase D, also known as prolidase, which regulates the recycling of proline) can result in SLE in a fraction of patients (96). Variants in MAN2B1 (encoding lysosomal alpha-d-mannosidase) can cause lysosomal α-mannosidosis and have been found in four patients with SLE (9799), one of which also carried an SLC7A7 variant (99). If proven causative, these genetic alterations reveal a complex relationship between metabolic disruptions and autoimmunity that require mechanistic understanding.

Implications for diagnosis and treatment

The increasing use of genomic sequencing in healthcare is assisting us in identifying an increasing number of potentially disease-causing genetic variations. The value of these findings depends on our ability to discern whether they are merely associated with disease or truly disease causing. Definitive genomic diagnosis holds significant implications for healthcare providers, patients, and their families as it can lead to a medical diagnosis, ending the diagnostic odyssey, provide families with severe hereditary diseases reproductive options, and guide the use of precision therapies. To aid in the interpretation of genetic findings, the National Institutes of Health established the ClinGen consortium in 2013 (100) with the objective of developing a reliable public resource that refines and establishes standards for evaluating the likelihood of a gene, and specific variants in each gene, being disease-causing and thus diagnostic. In 2022, a working group focusing on rheumatological autoimmune diseases was established, and in 2023 this group includes a gene curation expert panel (GCEP) with experts from multiple institutions that specifically focus on lupus (4). This effort is expected to lead to a refinement in the diagnosis and treatment of SLE, particularly early-onset forms.

A list of validated SLE-causing genes by the GCEP panel over the next two years will inform panel gene analysis of whole exome sequencing indicated for individuals with particularly severe or early onset SLE. Rare coding variants in validated lupus-causing genes can be classified according to ACMG guidelines (101). For those variants of unknown significance that do not reach the “likely pathogenic” or “pathogenic” classification criteria, assays that meet ClinGen guidelines for variant functional evaluation (102) will inform variant classification. There is a pressing need to develop such functional assays to test lupus variants, in ways that meet clinical and quality assurance standards.

While glucocorticoids, hydroxychloroquine, and immunosuppressive drugs continue to be the backbone of anti-inflammatory protocols in managing monogenic lupus, there is an increasing need for more targeted therapies to increase treatment efficacy. In some patients, lupus may be managed effectively with B cell-targeted treatments like Rituximab (103) and Belimumab (104), monoclonal antibodies that target CD20 and BAFF respectively. Recent studies, albeit in small numbers of patients, suggest that anti-CD19 chimeric antigen receptor (CAR) T cell therapy may outperform belimumab or rituximab, possibly through its ability to eliminate a greater number of tissue-resident B cells (105). An important role of type I IFN in disease progression suggests that medications designed to inhibit IFN-α/β receptors such as anifrolumab (106), may offer significant benefits to those suffering from mendelian SLE-like interferonopathies. Additional therapeutic options to decrease type I IFN signaling and formation of pathogenic ABCs cells include JAK inhibitors such as baricitinib and ruxolitinib (107, 108). Additionally, reverse transcriptase inhibitors, originally developed for HIV patients, have the potential to target endogenous retroviruses propose to be important SLE triggers (109) and reduce the IFN signature (110). Regular infusions of fresh frozen plasma (FFP) (111) and allogeneic hematopoietic stem cell transplantation (HSCT) may also prove to be effective options for patients with inherited complement deficiency or associated immunodeficiency.

Besides the generic treatment options described above, the question remains of how identification of functional rare variants in lupus genes may inform choice of therapy. For monogenic cases in which the variants occur within a gene confirmed to be lupus-causing or within genes clearly linked to others that are lupus causing, personalized treatment options may be possible. For example, patients identified to carry GoF TLR7 variants (45), or LoF UNC93B1 variants (52, 53) that disrupt degradative sorting of TLR7, may benefit from TLR7 inhibitors currently in phase II clinical trials. Additionally, mouse models of TLR7-driven lupus are dependent on IRAK4 (112), and several IRAK4 inhibitors are currently in phase I/II clinical trials (113), thus treatment with IRAK4 inhibitors may also be successful. Patients with pathogenic variants in specific DNases may benefit from administration of recombinant human DNase1 and DNase1L3 therapies. Dual-active DNases have recently been engineered and shown to be effective and degradation of NETs (114). Patients with TREX1 loss of function variants that lead to overactivation of the cGAS pathway may benefit from small molecule cGAS inhibitors. Given the reported cross-talks between these pathways (109), it is not unreasonable to speculate that a particular precision therapy may be effective across several pathways.

A greater challenge will arise in non-mendelian lupus cases. It is however possible that a significant fraction of lupus cases may be oligogenic in nature: two or a small number of rare variants with strong effects may be responsible for a large component of disease susceptibility, just as is being proposed for heart disease, hypercholesterolemia, and neurodegenerative disease (115117). These cases may benefit from combination therapies. A big unknown in the field is how many pathways are there to lupus, and whether many of the pathways today thought to be distinct interact or converge in one or a small number. At the rapid pace the field is moving, it is likely that interactions between some or most pathways described in this review will be revealed in the coming years.

Conclusion

SLE is a complex autoimmune disease, the understanding of which has experienced significant advancements in the last few years, particularly through whole exome/genome sequencing using next-generation sequencing technologies. Over 30 genes have been recognized as causes of monogenic SLE or SLE-like disease. This information is providing improved mechanistic insights that promise to aid in the identification of effective biomarkers and therapeutic strategies.

Key points.

  • Monogenic lupus, caused by variants in single genes, is a rare severe form of SLE usually manifesting during childhood.

  • Advancements in next-generation sequencing technologies have enabled the discovery of novel and ultra-rare variants causing monogenic SLE.

  • Although monogenic lupus constitutes a small fraction of lupus cases, the discovery of its causative variants greatly enhances understanding of disease pathogenesis illuminating potential therapeutic strategies.

  • Central to SLE pathogenesis are breakdown in nucleic acid tolerance, activation of the type I IFN pathway, and complement activation. Most gene mutations associated with monogenic lupus present with defects in these central mechanisms.

Acknowledgements

None.

Financial support and sponsorship

CV is funded by an LRA Global team award grant, a Royal Society Wolfson Fellowship, and a lupus insight prize, as well as the Francis Crick Institute (C2228), which receives its core funding from Cancer Research UK, the UK Medical Research Council, and the Wellcome Trust. YQ is funded by the National Natural Science Foundation of China (32170903 & 32270946).

Footnotes

Conflicts of interest

None.

References

* paper of special interest

** paper of outstanding interest

  • 1.Tsokos GC. Systemic Lupus Erythematosus. New England Journal of Medicine. 2011 doi: 10.1056/NEJMra1100359. [DOI] [PubMed] [Google Scholar]
  • 2.Deapen D, Escalante A, Weinrib L, et al. A revised estimate of twin concordance in systemic lupus erythematosus. Arthritis and rheumatism. 1992;35(3):311–8. doi: 10.1002/art.1780350310. [DOI] [PubMed] [Google Scholar]
  • 3.Block SR, Winfield JB, Lockshin MD, et al. Studies of twins with systemic lupus erythematosus. A review of the literature and presentation of 12 additional sets. The American journal of medicine. 1975;59(4):533–52. doi: 10.1016/0002-9343(75)90261-2. [DOI] [PubMed] [Google Scholar]
  • 4.Vinuesa CG, Shen N, Ware T. Genetics of SLE: mechanistic insights from monogenic disease and disease-associated variants. Nature reviews Nephrology. 2023;19(9):558–72. doi: 10.1038/s41581-023-00732-x. [DOI] [PubMed] [Google Scholar]
  • 5.Cirulli ET, Goldstein DB. Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nature Reviews Genetics. 2010;11(6):415–25. doi: 10.1038/nrg2779. [DOI] [PubMed] [Google Scholar]
  • 6.Manolio TA, Collins FS, Cox NJ, et al. Finding the missing heritability of complex diseases. Nature. 2009;461(7265):747–53. doi: 10.1038/nature08494. [** 5.6 are two excellent reviews about the contribution of rare variants to polygenic risk in complex diseases.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lo MS. Monogenic Lupus. Current rheumatology reports. 2016;18(12):71. doi: 10.1007/s11926-016-0621-9. [DOI] [PubMed] [Google Scholar]
  • 8.Omarjee O, Picard C, Frachette C, et al. Monogenic lupus: Dissecting heterogeneity. Autoimmunity reviews. 2019;18(10):102361. doi: 10.1016/j.autrev.2019.102361. [DOI] [PubMed] [Google Scholar]
  • 9.Costa-Reis P, Sullivan KE. Monogenic lupus: it’s all new! Current opinion in immunology. 2017;49:87–95. doi: 10.1016/j.coi.2017.10.008. [DOI] [PubMed] [Google Scholar]
  • 10.Sturfelt G, Truedsson L. Complement in the immunopathogenesis of rheumatic disease. Nature reviews Rheumatology. 2012;8(8):458–68. doi: 10.1038/nrrheum.2012.75. [DOI] [PubMed] [Google Scholar]
  • 11.Demirkaya E, Sahin S, Romano M, et al. New Horizons in the Genetic Etiology of Systemic Lupus Erythematosus and Lupus-Like Disease: Monogenic Lupus and Beyond. Journal of clinical medicine. 2020;9(3) doi: 10.3390/jcm9030712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Troldborg A, Steffensen R, Trendelenburg M, et al. Ficolin-3 Deficiency Is Associated with Disease and an Increased Risk of Systemic Lupus Erythematosus. Journal of clinical immunology. 2019;39(4):421–9. doi: 10.1007/s10875-019-00627-2. [DOI] [PubMed] [Google Scholar]
  • 13.Babaha F, Abolhassani H, Hamidi Esfahani Z, et al. A new case of congenital ficolin-3 deficiency with primary immunodeficiency. Expert review of clinical immunology. 2020;16(7):733–8. doi: 10.1080/1744666X.2020.1792779. [DOI] [PubMed] [Google Scholar]
  • 14.Ogden CA, deCathelineau A, Hoffmann PR, et al. C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells. The Journal of experimental medicine. 2001;194(6):781–95. doi: 10.1084/jem.194.6.781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Santer DM, Hall BE, George TC, et al. C1q deficiency leads to the defective suppression of IFN-alpha in response to nucleoprotein containing immune complexes. Journal of immunology (Baltimore, Md : 1950) 2010;185(8):4738–49. doi: 10.4049/jimmunol.1001731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Prodeus AP, Goerg S, Shen LM, et al. A critical role for complement in maintenance of self-tolerance. Immunity. 1998;9(5):721–31. doi: 10.1016/s1074-7613(00)80669-x. [DOI] [PubMed] [Google Scholar]
  • 17.Carter RH, Fearon DT. CD19: lowering the threshold for antigen receptor stimulation of B lymphocytes. Science (New York, NY) 1992;256(5053):105–7. doi: 10.1126/science.1373518. [DOI] [PubMed] [Google Scholar]
  • 18.Carroll MC. The complement system in B cell regulation. Molecular immunology. 2004;41(2-3):141–6. doi: 10.1016/j.molimm.2004.03.017. [DOI] [PubMed] [Google Scholar]
  • 19.Vinuesa CG, Linterman MA, Goodnow CC, Randall KL. T cells and follicular dendritic cells in germinal center B-cell formation and selection. Immunological reviews. 2010;237(1):72–89. doi: 10.1111/j.1600-065X.2010.00937.x. [DOI] [PubMed] [Google Scholar]
  • 20.Ling GS, Crawford G, Buang N, et al. C1q restrains autoimmunity and viral infection by regulating CD8(+) T cell metabolism. Science (New York, NY) 2018;360(6388):558–63. doi: 10.1126/science.aao4555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Leffler J, Martin M, Gullstrand B, et al. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. Journal of immunology (Baltimore, Md : 1950) 2012;188(7):3522–31. doi: 10.4049/jimmunol.1102404. [DOI] [PubMed] [Google Scholar]
  • 22.Han X, Xu T, Ding C, et al. Neuronal NR4A1 deficiency drives complement-coordinated synaptic stripping by microglia in a mouse model of lupus. Signal transduction and targeted therapy. 2022;7(1):50. doi: 10.1038/s41392-021-00867-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Caielli S, Wan Z, Pascual V. Systemic Lupus Erythematosus Pathogenesis: Interferon and Beyond. Annual review of immunology. 2023;41:533–60. doi: 10.1146/annurev-immunol-101921-042422. [** a comprehensive and insightful overview of the recent advancements in understanding the role of Interferon in pathogenesis of SLE.] [DOI] [PubMed] [Google Scholar]
  • 24.The type I interferonopathies: 10 years on. Nature Reviews Immunology. 2022 doi: 10.1038/s41577-021-00633-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sisirak V, Sally B, D’Agati V, et al. Digestion of Chromatin in Apoptotic Cell Microparticles Prevents Autoimmunity. Cell. 2016;166(1):88–101. doi: 10.1016/j.cell.2016.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Al-Mayouf SM, Sunker A, Abdwani R, et al. Loss-of-function variant in DNASE1L3 causes a familial form of systemic lupus erythematosus. Nature genetics. 2011;43(12):1186–8. doi: 10.1038/ng.975. [DOI] [PubMed] [Google Scholar]
  • 27.Hakkim A, Fürnrohr BG, Amann K, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(21):9813–8. doi: 10.1073/pnas.0909927107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yasutomo K, Horiuchi T, Kagami S, et al. Mutation of DNASE1 in people with systemic lupus erythematosus. Nature genetics. 2001;28(4):313–4. doi: 10.1038/91070. [DOI] [PubMed] [Google Scholar]
  • 29.Al-Mayouf SM, Alreefi HA, Alsinan TA, et al. Lupus manifestations in children with primary immunodeficiency diseases: Comprehensive phenotypic and genetic features and outcome. Modern rheumatology. 2021;31(6):1171–8. doi: 10.1080/14397595.2021.1886627. [DOI] [PubMed] [Google Scholar]
  • 30.Basu S, Sil A, Jindal AK, et al. A young boy with rash, arthritis, and developmental delay: Monogenic lupus due to DNASE2 gene defect. International journal of rheumatic diseases. 2023;26(12):2599–602. doi: 10.1111/1756-185X.14826. [DOI] [PubMed] [Google Scholar]
  • 31.Batlle-MasÓ L, Mensa-VilarÓ A, Solís-Moruno M, et al. Genetic diagnosis of autoinflammatory disease patients using clinical exome sequencing. Eur J Med Genet. 2020;63(5):103920. doi: 10.1016/j.ejmg.2020.103920. [DOI] [PubMed] [Google Scholar]
  • 32.Beltoise AS, Audouin-Pajot C, Lucas P, et al. [Familial chilblain lupus: Four cases spanning three generations] Annales de dermatologie et de venereologie. 2018;145(11):683–9. doi: 10.1016/j.annder.2018.07.014. [DOI] [PubMed] [Google Scholar]
  • 33.Linggonegoro DW, Song H, Jones KM, et al. Familial chilblain lupus in a child with heterozygous mutation in SAMHD1 and normal interferon signature. The British journal of dermatology. 2021;185(3):650–2. doi: 10.1111/bjd.20400. [DOI] [PubMed] [Google Scholar]
  • 34.Ramantani G, Häusler M, Niggemann P, et al. Aicardi-Goutières syndrome and systemic lupus erythematosus (SLE) in a 12-year-old boy with SAMHD1 mutations. Journal of child neurology. 2011;26(11):1425–8. doi: 10.1177/0883073811408310. [DOI] [PubMed] [Google Scholar]
  • 35.Ravenscroft JC, Suri M, Rice GI, et al. Autosomal dominant inheritance of a heterozygous mutation in SAMHD1 causing familial chilblain lupus. American journal of medical genetics Part A. 2011;155a(1):235–7. doi: 10.1002/ajmg.a.33778. [DOI] [PubMed] [Google Scholar]
  • 36.Zhao X, Li C, Li S, et al. SAMHD1 associates with inflammation and vasculitis in paediatric-onset systemic lupus erythematosus. Clinical and experimental rheumatology. 2022;40(9):1801–7. doi: 10.55563/clinexprheumatol/4k3gau. [DOI] [PubMed] [Google Scholar]
  • 37.Günther C, Kind B, Reijns MA, et al. Defective removal of ribonucleotides from DNA promotes systemic autoimmunity. The Journal of clinical investigation. 2015;125(1):413–24. doi: 10.1172/JCI78001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Batu ED. Monogenic systemic lupus erythematosus: insights in pathophysiology. Rheumatology international. 2018;38(10):1763–75. doi: 10.1007/s00296-018-4048-7. [DOI] [PubMed] [Google Scholar]
  • 39.Fiehn C. Familial chilblain lupus-what can we learn from type I interferonopathies? Current rheumatology reports. 2017;19:1–6. doi: 10.1007/s11926-017-0689-x. [DOI] [PubMed] [Google Scholar]
  • 40.Meyts I, Aksentijevich I. Deficiency of Adenosine Deaminase 2 (DADA2): Updates on the Phenotype, Genetics, Pathogenesis, and Treatment. Journal of clinical immunology. 2018;38(5):569–78. doi: 10.1007/s10875-018-0525-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Greiner-Tollersrud OK, Boehler V, Bartok E, et al. ADA2 is a lysosomal DNase regulating the type-I interferon response. bioRxiv. 2020 [Google Scholar]
  • 42.Skrabl-Baumgartner A, Plecko B, Schmidt WM, et al. Autoimmune phenotype with type I interferon signature in two brothers with ADA2 deficiency carrying a novel CECR1 mutation. Pediatric rheumatology online journal. 2017;15(1):67. doi: 10.1186/s12969-017-0193-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schepp J, Bulashevska A, Mannhardt-Laakmann W, et al. Deficiency of Adenosine Deaminase 2 Causes Antibody Deficiency. Journal of clinical immunology. 2016;36(3):179–86. doi: 10.1007/s10875-016-0245-x. [DOI] [PubMed] [Google Scholar]
  • 44.Xu L, Zhao J, Sun Q, et al. Loss-of-function variants in SAT1 cause X-linked childhood-onset systemic lupus erythematosus. Annals of the rheumatic diseases. 2022;81(12):1712–21. doi: 10.1136/ard-2022-222795. [* Newly discovered gene variants in SAT1 associated with monogenic lupus intriguingly establish a connection between SLE pathogenesis and polyamine metabolism, an area currently gainning significant attention in the study of intracelluar nucleic acid recognition.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Brown GJ, Cañete PF, Wang H, et al. TLR7 gain-of-function genetic variation causes human lupus. Nature. 2022;605(7909):349–56. doi: 10.1038/s41586-022-04642-z. [* First description of TLR7 gain-of-function variants as a cause of human monogenic SLE.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Van Eyck L, De Somer L, Pombal D, et al. Brief Report: IFIH1 Mutation Causes Systemic Lupus Erythematosus With Selective IgA Deficiency. Arthritis & rheumatology (Hoboken, NJ) 2015;67(6):1592–7. doi: 10.1002/art.39110. [DOI] [PubMed] [Google Scholar]
  • 47.Jeremiah N, Neven B, Gentili M, et al. Inherited STING-activating mutation underlies a familial inflammatory syndrome with lupus-like manifestations. The Journal of clinical investigation. 2014;124(12):5516–20. doi: 10.1172/JCI79100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.König N, Fiehn C, Wolf C, et al. Familial chilblain lupus due to a gain-of-function mutation in STING. Annals of the rheumatic diseases. 2017;76(2):468–72. doi: 10.1136/annrheumdis-2016-209841. [DOI] [PubMed] [Google Scholar]
  • 49.Peng J, Wang Y, Han X, et al. Clinical Implications of a New DDX58 Pathogenic Variant That Causes Lupus Nephritis due to RIG-I Hyperactivation. Journal of the American Society of Nephrology : JASN. 2023;34(2):258–72. doi: 10.1681/ASN.2022040477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wobma H, Shin DS, Chou J, Dedeoglu F. Dysregulation of the cGAS-STING Pathway in Monogenic Autoinflammation and Lupus. Frontiers in immunology. 2022;13:905109. doi: 10.3389/fimmu.2022.905109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Prasov L, Bohnsack BL, El Husny AS, et al. DDX58(RIG-I)-related disease is associated with tissue-specific interferon pathway activation. Journal of medical genetics. 2022;59(3):294–304. doi: 10.1136/jmedgenet-2020-107447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mishra H, Schlack-Leigers C, Lim EL, et al. Disrupted degradative sorting of TLR7 is associated with human lupus. Science immunology. 2024:eadi9575. doi: 10.1126/sciimmunol.adi9575. [DOI] [PubMed] [Google Scholar]
  • 53.Wolf C, Lim EL, Mokhtari M, et al. UNC93B1 variants underlie TLR7-dependent autoimmunity. Science immunology. 2024:eadi9769. doi: 10.1126/sciimmunol.adi9769. [ ** 52.53 newly discovered several variants in UNC93B1 leading to monogenic lupus, highlight the important role of abnormal degradation and increased activation of the TLR7 pathway in the pathogenesis of lupus. ] [DOI] [PubMed] [Google Scholar]
  • 54.Zhao K, Zhang M, Zhang L, et al. Intracellular osteopontin stabilizes TRAF3 to positively regulate innate antiviral response. Scientific reports. 2016;6:23771. doi: 10.1038/srep23771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.An J, Briggs TA, Dumax-Vorzet A, et al. Tartrate-Resistant Acid Phosphatase Deficiency in the Predisposition to Systemic Lupus Erythematosus. Arthritis & rheumatology (Hoboken, NJ) 2017;69(1):131–42. doi: 10.1002/art.39810. [DOI] [PubMed] [Google Scholar]
  • 56.Briggs TA, Rice GI, Daly S, et al. Tartrate-resistant acid phosphatase deficiency causes a bone dysplasia with autoimmunity and a type I interferon expression signature. Nature genetics. 2011;43(2):127–31. doi: 10.1038/ng.748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Xie C, Zhou H, Athanasopoulos V, et al. De Novo PACSIN1 Gene Variant Found in Childhood Lupus and a Role for PACSIN1/TRAF4 Complex in Toll-like Receptor 7 Activation. Arthritis & rheumatology (Hoboken, NJ) 2023;75(6):1058–71. doi: 10.1002/art.42416. [DOI] [PubMed] [Google Scholar]
  • 58.Zhang R, Yang W, Zhu H, et al. NLRC4 promotes the cGAS-STING signaling pathway by facilitating CBL-mediated K63-linked polyubiquitination of TBK1. Journal of medical virology. 2023;95(8):e29013. doi: 10.1002/jmv.29013. [DOI] [PubMed] [Google Scholar]
  • 59.Wang Q, Ye X, Zheng W, et al. NLRC4 gain-of-function variant is identified in a patient with systemic lupus erythematosus. Clinical immunology (Orlando, Fla) 2023;255:109731. doi: 10.1016/j.clim.2023.109731. [DOI] [PubMed] [Google Scholar]
  • 60.Schwickert TA, Tagoh H, Schindler K, et al. Ikaros prevents autoimmunity by controlling anergy and Toll-like receptor signaling in B cells. Nature immunology. 2019;20(11):1517–29. doi: 10.1038/s41590-019-0490-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Van Nieuwenhove E, Garcia-Perez JE, Helsen C, et al. A kindred with mutant IKAROS and autoimmunity. The Journal of allergy and clinical immunology. 2018;142(2):699–702.:e12. doi: 10.1016/j.jaci.2018.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hoshino A, Okada S, Yoshida K, et al. Abnormal hematopoiesis and autoimmunity in human subjects with germline IKZF1 mutations. The Journal of allergy and clinical immunology. 2017;140(1):223–31. doi: 10.1016/j.jaci.2016.09.029. [DOI] [PubMed] [Google Scholar]
  • 63.Dieudonné Y, Guffroy A, Vollmer O, et al. IKZF1 Loss-of-Function Variant Causes Autoimmunity and Severe Familial Antiphospholipid Syndrome. Journal of clinical immunology. 2019;39(4):353–7. doi: 10.1007/s10875-019-00643-2. [DOI] [PubMed] [Google Scholar]
  • 64.Mooney EC, Sahingur SE. The Ubiquitin System and A20: Implications in Health and Disease. Journal of dental research. 2021;100(1):10–20. doi: 10.1177/0022034520949486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tavares RM, Turer EE, Liu CL, et al. The ubiquitin modifying enzyme A20 restricts B cell survival and prevents autoimmunity. Immunity. 2010;33(2):181–91. doi: 10.1016/j.immuni.2010.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Duan R, Liu Q, Li J, et al. A De Novo Frameshift Mutation in TNFAIP3 Impairs A20 Deubiquitination Function to Cause Neuropsychiatric Systemic Lupus Erythematosus. Journal of clinical immunology. 2019;39(8):795–804. doi: 10.1007/s10875-019-00695-4. [DOI] [PubMed] [Google Scholar]
  • 67.Li G, Liu H, Li Y, et al. Genetic heterogeneity in Chinese children with systemic lupus erythematosus. Clinical and experimental rheumatology. 2021;39(1):214–22. doi: 10.55563/clinexprheumatol/zte897. [DOI] [PubMed] [Google Scholar]
  • 68.Kim HY, Song JY, Kim WI, et al. The First Case of an Infant with Familial A20 Haploinsufficiency in Korea. Journal of Korean medical science. 2020;35(30):e252. doi: 10.3346/jkms.2020.35.e252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nature reviews Immunology. 2005;5(5):375–86. doi: 10.1038/nri1604. [DOI] [PubMed] [Google Scholar]
  • 70.Zhang X, Bogunovic D, Payelle-Brogard B, et al. Human intracellular ISG15 prevents interferon-α/β over-amplification and auto-inflammation. Nature. 2015;517(7532):89–93. doi: 10.1038/nature13801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Al-Mayouf SM, Akbar L, AlEnazi A, Al-Mousa H. Autosomal Recessive ISG15 Deficiency Underlies Type I Interferonopathy with Systemic Lupus Erythematosus and Inflammatory Myositis. Journal of clinical immunology. 2021;41(6):1361–4. doi: 10.1007/s10875-021-01019-1. [DOI] [PubMed] [Google Scholar]
  • 72.Wu J, Wilson J, He J, et al. Fas ligand mutation in a patient with systemic lupus erythematosus and lymphoproliferative disease. The Journal of clinical investigation. 1996;98(5):1107–13. doi: 10.1172/JCI118892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Dasdemir S, Yildiz M, Celebi D, et al. Genetic screening of early-onset patients with systemic lupus erythematosus by a targeted next-generation sequencing gene panel. Lupus. 2022;31(3):330–7. doi: 10.1177/09612033221076733. [DOI] [PubMed] [Google Scholar]
  • 74.Kiykim A, Ogulur I, Baris S, et al. Potentially Beneficial Effect of Hydroxychloroquine in a Patient with a Novel Mutation in Protein Kinase Cδ Deficiency. Journal of clinical immunology. 2015;35(6):523–6. doi: 10.1007/s10875-015-0178-9. [DOI] [PubMed] [Google Scholar]
  • 75.Lei L, Muhammad S, Al-Obaidi M, et al. Successful use of ofatumumab in two cases of early-onset juvenile SLE with thrombocytopenia caused by a mutation in protein kinase C δ. Pediatric rheumatology online journal. 2018;16(1):61. doi: 10.1186/s12969-018-0278-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Nanthapisal S, Omoyinmi E, Murphy C, et al. Early-Onset Juvenile SLE Associated With a Novel Mutation in Protein Kinase C δ. Pediatrics. 2017;139(1) doi: 10.1542/peds.2016-0781. [DOI] [PubMed] [Google Scholar]
  • 77.Belot A, Kasher PR, Trotter EW, et al. Protein kinase cδ deficiency causes mendelian systemic lupus erythematosus with B cell-defective apoptosis and hyperproliferation. Arthritis and rheumatism. 2013;65(8):2161–71. doi: 10.1002/art.38008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Oliveira JB. The expanding spectrum of the autoimmune lymphoproliferative syndromes. Current opinion in pediatrics. 2013;25(6):722–9. doi: 10.1097/MOP.0000000000000032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bader-Meunier B, Cavé H, Jeremiah N, et al. Are RASopathies new monogenic predisposing conditions to the development of systemic lupus erythematosus? Case report and systematic review of the literature. Seminars in arthritis and rheumatism. 2013;43(2):217–9. doi: 10.1016/j.semarthrit.2013.04.009. [DOI] [PubMed] [Google Scholar]
  • 80.Hou Y, Wang L, Luo C, et al. Clinical characteristics of early-onset paediatric systemic lupus erythematosus in a single centre in China. Rheumatology (Oxford, England) 2023;62(10):3373–81. doi: 10.1093/rheumatology/kead086. [DOI] [PubMed] [Google Scholar]
  • 81.Li G, Li Y, Liu H, et al. Genetic heterogeneity of pediatric systemic lupus erythematosus with lymphoproliferation. Medicine. 2020;99(20):e20232. doi: 10.1097/MD.0000000000020232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Li GM, Liu HM, Guan WZ, et al. A mutation in PIK3CD gene causing pediatric systemic lupus erythematosus: A case report. Medicine. 2019;98(18):e15329. doi: 10.1097/MD.0000000000015329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Thouenon R, Moreno-Corona N, Poggi L, et al. Activated PI3Kinase Delta Syndrome-A Multifaceted Disease. Frontiers in pediatrics. 2021;9:652405. doi: 10.3389/fped.2021.652405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.He Y, Gallman AE, Xie C, et al. P2RY8 variants in lupus patients uncover a role for the receptor in immunological tolerance. The Journal of experimental medicine. 2022;219(1) doi: 10.1084/jem.20211004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Biggs CM, Keles S, Chatila TA. DOCK8 deficiency: Insights into pathophysiology, clinical features and management. Clinical immunology (Orlando, Fla) 2017;181:75–82. doi: 10.1016/j.clim.2017.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhang Yaoyuan, Morris Rhiannon, Brown Grant J, et al. Rare SH2B3 coding variants in lupus patients impair B cell tolerance and predispose to autoimmunity. The Journal of experimental medicine. 2024 doi: 10.1084/jem.20221080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Paç Kisaarslan A, Witzel M, Unal E, et al. Refractory and Fatal Presentation of Severe Autoimmune Hemolytic Anemia in a Child With the DNASE1L3 Mutation Complicated With an Additional DOCK8 Variant. Journal of pediatric hematology/oncology. 2021;43(3):e452–e6. doi: 10.1097/MPH.0000000000001780. [DOI] [PubMed] [Google Scholar]
  • 88.Seo E, Lee BH, Lee JH, et al. Hematopoietic stem cell transplantation in an infant with dedicator of cytokinesis 8 (DOCK8) deficiency associated with systemic lupus erythematosus: A case report. Medicine. 2021;100(13):e20866. doi: 10.1097/MD.0000000000020866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jouhadi Z, Khadir K, Ailal F, et al. Ten-year follow-up of a DOCK8-deficient child with features of systemic lupus erythematosus. Pediatrics. 2014;134(5):e1458–63. doi: 10.1542/peds.2013-1383. [DOI] [PubMed] [Google Scholar]
  • 90.Liphaus BL, Caramalho I, Rangel-Santos A, et al. LRBA deficiency: a new genetic cause of monogenic lupus. Annals of the rheumatic diseases. 2020;79(3):427–8. doi: 10.1136/annrheumdis-2019-216410. [DOI] [PubMed] [Google Scholar]
  • 91.Lo B, Zhang K, Lu W, et al. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science (New York, NY) 2015;349(6246):436–40. doi: 10.1126/science.aaa1663. [DOI] [PubMed] [Google Scholar]
  • 92.Jang E, Kim UK, Jang K, et al. Bach2 deficiency leads autoreactive B cells to produce IgG autoantibodies and induce lupus through a T cell-dependent extrafollicular pathway. Experimental & molecular medicine. 2019;51(12):1–13. doi: 10.1038/s12276-019-0352-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Zhou L, Sun G, Chen R, et al. An early-onset SLE patient with a novel paternal inherited BACH2 mutation. Journal of clinical immunology. 2023;43(6):1367–78. doi: 10.1007/s10875-023-01506-7. [DOI] [PubMed] [Google Scholar]
  • 94.De Ravin SS, Naumann N, Cowen EW, et al. Chronic granulomatous disease as a risk factor for autoimmune disease. The Journal of allergy and clinical immunology. 2008;122(6):1097–103. doi: 10.1016/j.jaci.2008.07.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Noguchi A, Takahashi T. Overview of symptoms and treatment for lysinuric protein intolerance. Journal of human genetics. 2019;64(9):849–58. doi: 10.1038/s10038-019-0620-6. [DOI] [PubMed] [Google Scholar]
  • 96.Butbul Aviel Y, Mandel H, Avitan Hersh E, et al. Prolidase deficiency associated with systemic lupus erythematosus (SLE): single site experience and literature review. Pediatric rheumatology online journal. 2012;10(1):18. doi: 10.1186/1546-0096-10-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Lipiński P, Różdżynska-Świątkowska A, Iwanicka-Pronicka K, et al. Long-term outcome of patients with alpha-mannosidosis - A single center study. Molecular genetics and metabolism reports. 2022;30:100826. doi: 10.1016/j.ymgmr.2021.100826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Urushihara M, Kagami S, Yasutomo K, et al. Sisters with alpha-mannosidosis and systemic lupus erythematosus. Eur J Pediatr. 2004;163(4-5):192–5. doi: 10.1007/s00431-004-1404-2. [DOI] [PubMed] [Google Scholar]
  • 99.Tirosh I, Spielman S, Barel O, et al. Whole exome sequencing in childhood-onset lupus frequently detects single gene etiologies. Pediatric rheumatology online journal. 2019;17(1):52. doi: 10.1186/s12969-019-0349-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Rehm HL, Berg JS, Brooks LD, et al. ClinGen--the Clinical Genome Resource. The New England journal of medicine. 2015;372(23):2235–42. doi: 10.1056/NEJMsr1406261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24. doi: 10.1038/gim.2015.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Brnich SE, Abou Tayoun AN, Couch FJ, et al. Recommendations for application of the functional evidence PS3/BS3 criterion using the ACMG/AMP sequence variant interpretation framework. Genome Medicine. 2019;12(1):3. doi: 10.1186/s13073-019-0690-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Faustini F, Sippl N, Stålesen R, et al. Rituximab in Systemic Lupus Erythematosus: Transient Effects on Autoimmunity Associated Lymphocyte Phenotypes and Implications for Immunogenicity. Frontiers in immunology. 2022;13:826152. doi: 10.3389/fimmu.2022.826152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ramsköld D, Parodis I, Lakshmikanth T, et al. B cell alterations during BAFF inhibition with belimumab in SLE. EBioMedicine. 2019;40:517–27. doi: 10.1016/j.ebiom.2018.12.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mackensen A, Müller F, Mougiakakos D, et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nature medicine. 2022;28(10):2124–32. doi: 10.1038/s41591-022-02017-5. [DOI] [PubMed] [Google Scholar]
  • 106.Morand EF, Furie R, Tanaka Y, et al. Trial of Anifrolumab in Active Systemic Lupus Erythematosus. The New England journal of medicine. 2020;382(3):211–21. doi: 10.1056/NEJMoa1912196. [DOI] [PubMed] [Google Scholar]
  • 107.Ma L, Peng L, Zhao J, et al. Efficacy and safety of Janus kinase inhibitors in systemic and cutaneous lupus erythematosus: A systematic review and meta-analysis. Autoimmunity reviews. 2023:103440. doi: 10.1016/j.autrev.2023.103440. [DOI] [PubMed] [Google Scholar]
  • 108.Dai D, Gu S, Han X, et al. The transcription factor ZEB2 drives the formation of age-associated B cells. Science (New York, NY) 2024;383(6681):413–21. doi: 10.1126/science.adf8531. [ * Identification of ZEB2 as the trasncription factor that drives pathogenic lupus B cells, and its regulation by the JAK-STAT pathway, providing an additional rationale for inhibtors of this pathway to treat SLE. ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Vinuesa CG, Grenov A, Kassiotis G. Innate virus-sensing pathways in B cell systemic autoimmunity. Science (New York, NY) 2023;380(6644):478–84. doi: 10.1126/science.adg6427. [ * This review explains how anti-viral immunity operates at the verge of autoimmunity, with defects in the same nucleic acid sensing proteins causing severe viral infections when underactive, or autoimmunity when overactive. ] [DOI] [PubMed] [Google Scholar]
  • 110.Crow YJ, Vanderver A, Orcesi S, et al. Therapies in Aicardi-Goutières syndrome. Clinical and experimental immunology. 2014;175(1):1–8. doi: 10.1111/cei.12115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zecevic M, Minic A, Pasic S, et al. Case Report: Early Onset Systemic Lupus Erythematosus Due to Hereditary C1q Deficiency Treated With Fresh Frozen Plasma. Frontiers in pediatrics. 2021;9:756387. doi: 10.3389/fped.2021.756387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Celhar T, Lu HK, Benso L, et al. TLR7 Protein Expression in Mild and Severe Lupus-Prone Models Is Regulated in a Leukocyte, Genetic, and IRAK4 Dependent Manner. Frontiers in immunology. 2019;10:1546. doi: 10.3389/fimmu.2019.01546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Parrondo RD, Iqbal M, Von Roemeling R, et al. IRAK-4 inhibition: emavusertib for the treatment of lymphoid and myeloid malignancies. Frontiers in immunology. 2023;14:1239082. doi: 10.3389/fimmu.2023.1239082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Englert H, Göbel J, Khong D, et al. Targeting NETs using dual-active DNase1 variants. Frontiers in immunology. 2023;14:1181761. doi: 10.3389/fimmu.2023.1181761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gifford CA, Ranade SS, Samarakoon R, et al. Oligogenic inheritance of a human heart disease involving a genetic modifier. Science (New York, NY) 2019;364(6443):865–70. doi: 10.1126/science.aat5056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kirola L, Mukherjee A, Mutsuddi M. Recent Updates on the Genetics of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Mol Neurobiol. 2022;59(9):5673–94. doi: 10.1007/s12035-022-02934-z. [DOI] [PubMed] [Google Scholar]
  • 117.Tada H, Nohara A, Kawashiri MA. Monogenic, polygenic, and oligogenic familial hypercholesterolemia. Curr Opin Lipidol. 2019;30(4):300–6. doi: 10.1097/QCO.0000000000000563. [DOI] [PubMed] [Google Scholar]

RESOURCES