Abstract
Mouse models of SLE have been indispensable tools to study disease pathogenesis, to identify genetic susceptibility loci and targets for drug development, and for preclinical testing of novel therapeutics. Recent insights into immunological mechanisms of disease progression have boosted a revival in SLE drug development. Despite promising results in mouse studies, many novel drugs have failed to meet clinical end points. This is probably because of the complexity of the disease, which is driven by polygenic predisposition and diverse environmental factors, resulting in a heterogeneous clinical presentation. Each mouse model recapitulates limited aspects of lupus, especially in terms of the mechanism underlying disease progression. The main mouse models have been fairly successful for the evaluation of broad-acting immunosuppressants. However, the advent of targeted therapeutics calls for a selection of the most appropriate model(s) for testing and, ultimately, identification of patients who will be most likely to respond.
Keywords: systemic lupus erythematosus, SLE, mouse models, SLE treatment, SLE modelling, NZB/W mouse, MRL/lpr mouse, TLR7, IFN
Mouse models are indispensable tools to study human SLE.
Mouse models recapitulate specific elements of SLE, particularly with regard to the mechanism of disease.
Selection of the most appropriate model(s) for future testing of targeted SLE therapeutics is essential.
Introduction
SLE is a complex autoimmune disease with an extremely heterogeneous clinical presentation, which reflects the multiple roles of the environment, genetics and immune response in disease initiation and progression [1]. Variability in disease manifestation and severity makes it extremely challenging to study in clinical trials, especially in terms of selecting the patient population who will be most likely to respond to the treatment under investigation [2].
Murine models of disease represent genetically homogeneous populations to study the initiation and the progressive pathogenesis at the local, peripheral and end-organ stage [3]. They provide a much faster system for therapeutic screening, because mice reach 50% mortality due to GN at 5–9 months of age [4, 5]. Moreover, they allow for examination in the absence of any therapy, which is a major caveat of studying samples from SLE patients, who still take chronic doses of immunosuppressants even when in remission [6]. Additionally, they allow for easy assessment of drug combinations with the aim of reducing individual doses and side effects of high-dose monotherapy.
The limitations to use of murine models are the increasing costs, the longevity of projects and the scientific expertise to design, fulfil, analyse and interpret results to ascertain meaningful data applicable to human disease. In this review, we briefly discuss the commonalities and differences of the most commonly used mouse strains in lupus research and highlight how they have provided a meaningful path forward for therapeutic intervention.
Main mouse models
Defining whether a system is a good model of disease requires an analysis of its requirements. A hallmark feature of human SLE is the presence of ANAs, anti-dsDNA antibodies and anti-RNA or RNA-associated antibodies. Clinical manifestations include GN, arthritis, heart disease, cutaneous lesions and neurological symptoms [1]. Likewise, as each patient presents with a unique phenotype, mouse models can recapitulate only limited features of the disease (Table 1).
Table 1.
Lupus models | Main genetic components | Related human genetic associations | Immunological characteristics | Model used to assess | |
---|---|---|---|---|---|
Spontaneous |
|
|
Ccr2, FcgRIIA, FcgRIIIA, FcgRIIIb, PARP, CRP/SAP, kallikrein genes, possibly SLAMF genes |
|
|
MRL/lpr | Fas mutation (lpr) | Fas/FasL polymorphisms |
|
|
|
|
|
|
|
|
|
Accelerated |
|
Strain dependent |
|
|
|
Induced | Pristane induced (BALB/c, C57BL/6, DBA/1, SJL) | Pristane induces lupus in mice lacking genetic predisposition |
|
|
|
Spontaneous New Zealand black×white F1 (NZB/W), Murphy Roths Large/lymphoproliferative (MRL/lpr) and BXSB mouse models have been extensively used for studying immunological mechanisms and therapeutic targets over the last 40 years [5]. All three models exhibit serum ANAs and develop GN according to a strain- and sex-specific time line [4, 5]. Based on their specificities, these models can be used to examine the role of different genes and pathways, cellular dependency in disease progression and therapeutic targeting. Additionally, IFNα-dependent models and Toll-like receptor 7 (TLR7)-associated strains are gaining in importance, based on the extensive data supporting these pathways in the pathogenesis of SLE [7–9].
MRL/lpr
MRL/lpr mice have a loss-of-function lymphoproliferation (lpr) mutation within the gene encoding Fas, a cell-surface protein that mediates apoptosis [10]. They are characterized by lymphoproliferation, enlarged lymph nodes (lymphadenopathy) and GN, and between 25 and 75% mice develop arthritis. Serologically, they display hyperimmunoglobulinaemia, high ANAs, high anti-dsDNA antibodies and anti-small nuclear ribonucleoprotein (sn-RNP) antibodies. Male and female MLR/lpr mice are equally affected [4, 5]. Lymphadenopathy and splenomegaly are attributable to expansion of an unusual double-negative CD4−CD8−CD3+ B220+ T cell population [11]. Aside from examining the mechanisms of autoantibody production and renal failure, MRL/lpr mice are also used to examine cutaneous and neurological aspects of lupus, in contrast with other strains [12, 13].
Human relevance
MRL/lpr mice recapitulate many features of lupus; however, massive lymphadenopathy is not typical of human disease. Nonetheless, several recent studies reported an association of Fas and Fas ligand polymorphisms with the susceptibility to SLE, and increased double-negative T cells have been found in the periphery and in the kidneys of SLE patients [14–17].
IFN dependency
Pre-autoimmune MRL/lpr mice do not show evidence of elevated IFN-induced genes (i.e. IFN signature) [18]. IFN receptor (IFNAR) deficiency enhanced the disease, and anti-IFNAR antibody treatment did not mediate any long-term effects in this model [19, 20]. Consequently, MRL/lpr mice are not appropriate for studying the role of type I IFN in lupus.
NZB/W
The disease in New Zealand black×white F1 hybrid (NZB/W) mice has a strong female bias, and it is characterized by lymphadenopathy, splenomegaly, increased concentrations of ANA and anti-dsDNA antibodies and IC-mediated GN [4, 5]. NZB/W mice are also used as a model of lupus-related cardiovascular disease [21].
Crossing and selective inbreeding generated several New Zealand mixed (NZM) strains, with diverse phenotypic traits and variability in penetrance, severity, onset and gender bias [22]. The NZM2410 strain rapidly develops severe GN in both female and male mice, whereas GN in NZM2328 mice is female biased [22, 23].
Human relevance
Arguably, the most important contribution of the genetic studies in NZB/W-congenic derivatives was the identification of the NZM2410-derived Sle1 and NZB-derived Nba2 locuses, which are responsible for the production of autoantibodies [24]. Sle1 and Nba2 overlap in the telomeric region of chromosome 1, which encodes members of the FcγR, SLAM and IFN-inducible (Ifi) receptor families [25–30]. This region has a human syntenic equivalent on chromosome 1, 1q21–44, which has been associated with SLE in human linkage studies [31–33]. Human gene associations include Cr2 [34, 35], FcγRIIA, FcγRIIIA and FcγRIIIb [36–38], PARP [39] and CRP/SAP [40, 41]. SLAM family members Ly108 and CD84 have been identified as disease causative in mice, but they may be less significant in human SLE [29, 42]. NZM2328-derived susceptibility locuses associated with GN, Cgnz1 and Agnz1, are also located on the distal region of chromosome 1, overlapping with Sle1 [23, 43]. Cgnz1 has a nearly identical homologous region in the human genome; however, further studies are needed to identify possible susceptibility genes [43].
NZM2410-derived Sle3 on chromosome 7 is responsible for generalized T cell activation and development of nephritis [44, 45]. The kallikrein genes within this region were associated with nephritis in both mice and humans [46]. NZM2410-derived region Sle2, responsible for the expansion of autoreactive B cells, and NZM2328-derived region Adnz1, responsible for autoantibody production, are located on mouse chromosome 4 [47, 48]. These regions are under investigation to identify novel susceptibility genes.
IFN dependency
Pre-autoimmune NZB/W mice display elevated IFN-regulated gene expression in the spleen [18]. The IFN signature has also been observed in myeloid dendritic cells from the triple-congenic NZM2410-derived Sle123 strain [49]. Additionally, IFNAR deficiency has been shown to reduce disease in NZM2328 and NZM2328-derived B6.Nba2 mice [50, 51].
BXSB and associated strains with TLR7 upregulation
BXSB mice develop a rapid-onset severe disease in males [4, 5]. The male bias is attributable to the presence of the Y-autoimmune accelerator (Yaa) locus, which arose from an X to Y chromosome translocation [52, 53]. This doubled the genomic copy number and therefore the expression of a number of genes, including TLR7. TLR7 is crucial for the severe aspects of pathogenesis in this model [54–56]. Yaa can also accelerate disease in MRL, NZW and NZB lupus-prone mice [57, 58]. Likewise, a 2-fold upregulation of TLR7 on a B6.Sle1 mild autoimmune-prone background (Sle1.Tg7) or larger increases in expression (>4- to 8-fold; TLR7.Tg) on a non-autoimmune-prone background are sufficient to drive severe disease [54, 59]. BXSB susceptibility loci are required for the development of the disease, because Yaa is not sufficient to cause lupus in mice that lack an autoimmune genetic predisposition [57, 60]. BXSB-derived loci, designated Bxsb1–6, are present on various chromosomes and many overlap with known NZB/W-derived regions [61, 62].
A unique feature of the BXSB strain is the TLR7-dependent expansion of circulating monocytes (monocytosis) [56]. An increased proportion of monocytes, especially non-classical CD14+ CD16++, has also been observed in SLE patients [63, 64]. Additionally, strains with increased TLR7 expression have reduced splenic marginal zone cells and an expansion of T follicular helper cells and myeloid cells [52–54, 56, 59]. Aged mice develop GN and show increased leucocyte infiltration into the kidney, particularly CD11b+ myeloid cells [53–56, 59]. TLR7-associated strains have rarely been used to assess drug targets, possibly because of their male gender bias, which is unlike human disease. However, they have provided important insight into the immune mechanisms driving end-organ pathology.
Human relevance
The Bxsb3 locus overlaps with Sle1 and Nba2 and has been associated with autoantibody production and GN [61, 62]. The genes in this region that might be of relevance in human SLE are FcγgRII and Ifi202 [65]. Human translocations from the X to Y chromosome have not been found; nonetheless, SLE is more prevalent in men who have an additional X chromosome [66]. Increased TLR7 gene copies and two single nucleotide polymorphisms, rs179008 and rs3853839, have been associated with SLE in different ethnicities [67, 68]. Signalling pathways downstream of TLR7 can also be affected, as exemplified by IRF5, which is strongly associated with SLE susceptibility [69, 70]. Elevated TLR7 expression appears to be a common feature of peripheral blood mononuclear cells from SLE patients; it can correlate with IFNα expression and can be induced by IFNα itself in several immune cells [71]. Immunological studies of human SLE have also shown a role of TLR7 in neutrophil extracellular trap cell death and generation of anti-snRNP antibodies [72, 73].
IFN dependency
The IFN signature has been observed in the kidneys of BXSB mice [74]. Treatment with an anti-IFNAR antibody was effective, particularly if started at early stages [20]. Thus, BXSB might represent a model for elucidating the role of type I IFN in the early stages of the disease.
IFNα-dependent/driven mouse models
A strong IFN signature in the peripheral blood can be induced by pristane and other hydrocarbons in non-autoimmune-prone mice, such as BALB/c, C57BL/6 and DBA/1 [75]. The disease in these mice is TLR7 and IFN dependent and characterized by GN, ANAs, anti-dsDNA antibodies, anti-snRNP antibodies and arthritis [75].
Additionally, IFNα can be used to induce rapid-onset, severe lupus in spontaneous models, such as NZB/W, B6.Sle123 and NZM2328. These mice represent a reliable but stringent model to evaluate new drugs, because they do not respond well to standard therapies [76].
Evaluation of standard-of-care SLE therapeutics in mouse models
To date, only a handful of drugs have been approved for the treatment of SLE. In the 1950s, the US Food and Drug Administration approved aspirin, CSs and the antimalarial drug HCQ as non-specific treatments for SLE. These drugs were not approved following clinical trials, but based on clinical experience and eminence-based intuition. It took nearly 60 years of research before the approval of the next therapeutic and first targeted biologic, belimumab, in 2011 [77]. Additionally, several off-label agents were introduced to SLE therapy, predominantly systemic immunosuppressants such as CYC, MTX and MMF [77]. Despite being the pillars of SLE treatment [78], their mechanisms of action are not completely understood. NZB/W and MRL/lpr models have been extensively used to study these mechanisms and to evaluate side effects, dosage regimens and response to treatment, especially the ability to delay or prevent renal disease [5].
CYC
CYC is widely used as a chemotherapeutic and immunosuppressant with remarkable immunodepletive properties [79]. The first murine lupus studies involving CYC began in the late 1960s using the NZB/W mouse model. In these studies, CYC decreased autoantibody production and repressed the progression of LN without reversing the existing abnormalities [80, 81]. Protection from severe GN was achieved with long-term high-dose CYC and correlated with decreased anti-DNA antibody levels [82]. Short courses of intermittent pulse CYC did not achieve sustained immunosuppression [83, 84]. The efficacy of CYC has been confirmed in the MRL/lpr model; it prolonged survival, decreased arthritis and nephritis, reduced adenopathy and splenomegaly, reduced antibody levels and normalized T and B cells [85, 86].
Immunological studies on NZB/W mice have shown that CYC therapy depletes dividing, short-lived plasmablasts, but does not delete long-lived plasma cells efficiently [87]. These persist in survival niches, which are provided by the bone marrow and inflamed tissues, and might explain the resistance to immunosuppressive therapy observed in both mice and humans [88–90]. Fifty per cent of SLE patients show persistent active nephritis despite the therapy showing apparent clinical response [91].
Long-term CYC therapy is more efficient; however, it is associated with more side effects. Prolonged administration of CYC, especially high doses, increases the incidence of neoplasms in NZB/W mice [92, 93]. Long-term CYC (>1 year) in humans is also carcinogenic, causing most frequently bladder cancer, secondary acute leukaemia and skin cancer [79]. Owing to its toxicity, CYC is commonly used at a lower dose in combination with other drugs. In the 1970s, Steinberg et al. went on to study the combination of CYC, AZA and methylprednisolone (Mp) in NZB/W mice [94]. Prevention of GN was achieved either by intermittent high doses of CYC or by daily low doses of a combination of CYC, AZA and Mp. These treatments were beneficial only when started early or in older mice with mild renal disease [94]. These studies provided the basis for human trials, which concluded that the combination of an immunosuppressant and low-dose prednisone was superior to treatment with a high dose of CSs alone at preserving renal function [95, 96]. However, for most of the treatment regimens, the effect was evident only after 5–7 years from the initiation of the trial [95].
Methylprednisolone, prednisolone and prednisone
CSs have been extensively used to suppress inflammation in a variety of immune-mediated diseases [97]. CS therapy, especially high dose and long term, is associated with many complications, including weight gain, hypertension, atherosclerosis, diabetes, peptic ulcer, skin atrophy, acne vulgaris and increased risk for infections [97]. Despite their many side effects and unclear mechanism of action, CSs (in combination with HCQ, CYC, MTX or MMF) remain the preferred induction therapy for almost all clinical presentations of lupus [78]. CSs in combined regimens have been extensively studied in murine lupus; however, very few studies have addressed the effects and mechanism of action of steroids alone. Mp administered to NZB/W mice at the onset of nephritis preserved the glomerular structure and function by decreasing the amount of IgG, IgM and C3 deposits. This was associated with lower plasma concentration of IgG, but not of anti-DNA antibodies, C3 and C1q-reactive materials [98]. Mp also decreased proteinuria by preserving glomerular permeability and improved survival [99]. CSs might preserve renal function through the inhibition of nuclear factor-κB (NF-κB) activity, which has been implied in LN pathogenesis [100, 101]. Mp has been shown to inhibit the lipopolysaccharide-induced activation of NF-κB in the kidneys of MRL/lpr mice [102]. Additionally, prednisolone inhibited the expression of many NF-κB-inducible genes in the glomeruli of MRL/lpr mice, such as adhesion molecules, chemokines and their receptors and proteins involved in antigen presentation [103, 104]. Both prednisolone and methylprednisolone attenuated the expression of extracellular matrix components in the kidneys of MRL/lpr and NZB/W mice, respectively [103, 105].
MMF
MMF is an immunosuppressive drug used to reduce acute and chronic transplant rejection. In SLE, it is most commonly used together with CSs in the induction therapy of LN [78]. When administered to NZB/W mice, MMF (60 and 200 mg/kg/day) reduced proteinuria, albuminuria and blood urea nitrogen concentrations, decreased autoantibody production and prolonged survival [106, 107]. Low doses of MMF (30 mg/kg/day) were effective in diminishing glomerular lesions and promoted qualitative changes in autoantibody production, lowering specifically IgG2a antibodies, but did not affect serum IgG or anti-dsDNA antibody levels [108]. Likewise, in MRL/lpr mice, 90 mg/kg/day MMF efficiently reduced albuminuria and GN and caused less immunoglobulin and C3 deposition in the glomeruli, but did not diminish antibody formation [109]. However, when MMF was given at 100 mg/kg/day, it reduced serum levels of antibodies in both NZB/W and MRL/lpr mice [108, 110]. MMF at 300 mg/kg/day also effectively abrogated LN development in the IFNα-accelerated NZB/W model and led to a decrease in the number of antibody-secreting cells in the spleen, but did not affect serum levels of anti-dsDNA antibody [111].
In the MRL/lpr renal cortex, MMF inhibits expression of inducible nitric oxide synthase, at least in the initial stages of disease [112, 113]. In contrast with CSs, it does not affect the NF-κB pathway [112, 114]. In the kidneys of NZB/W mice, MMF has been found to reduce the activation of protein kinase C, reduce fibronectin expression and reduce the expression of urokinase receptor in podocytes [115, 116].
Antimalarial agents
Use of the antimalarial agents quinacrine, chloroquine (CQ) and HCQ in SLE has recently been extensively reviewed [117, 118]. Their principal modes of action are thought to be interference with lysosomal acidification and inhibition of TLR7/9 through binding of nucleic acids [118].
Two studies have assessed HCQ for the treatment of lupus-associated endothelial dysfunction in NZB/W mice. Long-term treatment with 10 mg/kg/day HCQ by oral gavage resulted in reduced hypertension, reduced endothelial dysfunction and less damage of the heart and kidneys, without altering anti-dsDNA antibody levels [119]. Early HCQ treatment with a lower dose (3 mg/kg/day) also reduced endothelial dysfunction, but did not decrease the degree of nephritis [21]. These effects have been attributed to anti-oxidative properties of HCQ, but further studies are needed to elucidate the exact mechanism. The potential for HCQ to treat lupus-related skin lesions has been evaluated in the MRL/lpr model, showing efficacy and safety [120].
Mouse models for the screening of targeted therapeutics
B cell-targeted therapies
B cells produce antibodies (including autoantibodies), cytokines and chemokines and are crucial for normal humoral immune function. They represent an obvious and valid drug target in autoimmune diseases, as recently proved by the approval of belimumab for SLE treatment [121].
Belimumab
Belimumab is a human monoclonal antibody that binds and neutralizes the B cell-activating factor, B-lymphocyte stimulator (BLyS, commonly known as BAFF). BAFF and its homologue APRIL (a proliferation-inducing ligand) bind to receptors that are expressed on B cells at different stages of maturation [122].
Mouse experiments played an indispensable part in elucidating the role of BAFF and establishing it as an effective target for SLE treatment [121, 122]. Administration of recombinant BAFF to non-lupus mice results in elevated immunoglobulin production [123]. BAFF-transgenic (BAFF-Tg) mice develop SLE-like manifestations, and introduction of a BAFF transgene into autoimmune-prone B6.Sle1 or B6.Nba2 mice accelerates the development of GN [124, 125]. BAFF inhibition reduces nephritis and prolongs survival in multiple mouse models: NZM2410, NZB/W and BXSB [126–128]. Additionally, genetic ablation of BAFF prevents IFNα-dependent acceleration of GN, suggesting that therapeutic targeting of BAFF could be successful in patients with an IFNα signature [129].
Mouse models are also being used to elucidate the exact mechanism of action of belimumab. Older studies suggested that the blockade of BAFF would delete strongly self-reactive B cells and thus limit the autoimmune response [130]. However, new data suggest that BAFF blockade prevents signalling through its receptor transmembrane activator, calcium modulator, and cyclophilin ligand interactor, resulting in complete protection from autoantibody production without an extensive reduction in the number of B cells [131]. This could partly explain the controversial efficacy of B cell-depleting treatments, such as rituximab (anti-CD20 monoclonal antibody) [132].
Anti-CD20 B cell-depleting therapies
In humanized MRL/lpr mice, which express hCD20, anti-CD20 therapy effectively reduces B cell numbers and leads to amelioration of the disease, albeit only when used at high doses and for a prolonged time [133]. Unusually high doses (1–10 mg per mouse) have been used because of resistance to depletion that was later attributed to impaired IgG-mediated phagocytosis [134]. NZB/W mice also display less effective B cell depletion compared with C57BL/6 mice that increases with age and disease severity [135]. Nonetheless, a short course (4 weeks) of anti-CD20 therapy (10 mg/kg) in young NZB/W mice delayed the onset of the disease and delayed the development of nephritis in mice with advanced disease without decreasing anti-dsDNA antibodies [135]. Reduction of autoantibodies and prevention of renal injury and hypertension were achieved only after long-term B cell depletion [136].
Anti-CD20 B cell depletion can be further enhanced by concomitant BAFF blockade, leading to reduction of autoantibodies, reduced kidney disease and prolonged survival [135, 137]. Dual anti-CD20 and anti-BAFF therapy has also been effective in the IFNα- or pristane-accelerated NZB/W mice, particularly in attenuating renal injury [137].
Despite the promising results of preclinical trials, especially in NZB/W mice, clinical trials with the anti-CD20 mAb rituximab did not meet the chosen end points [132]. However, the cumulative data from randomized control trials, open clinical trials and cohort studies have shown that rituximab is a safe and effective treatment for non-renal manifestations of SLE, such as arthritis and thrombocytopaenia, and is currently being used off-label [132, 138]. Based on mouse data, the dual anti-BAFF/anti-CD20 therapy might be useful to treat renal manifestations of SLE, especially in the context of IFN signature.
T cell CTLA-4 co-stimulation blockade
Cytotoxic T-lymphocyte antigen 4 (CTLA-4 or CD152) is an inhibitory protein on the surface of T cells that binds CD80/CD86 on antigen-presenting cells. By inhibiting CD28-mediated T cell activation, CTLA-4 plays a role in regulation of central tolerance and prevention of self-reactivity [139]. The same effect is mediated by the recombinant fusion protein CTLA-4-Ig (abatacept). Abatacept has been approved for the treatment of RA and has shown promising results in murine LN; however, it failed in human SLE trials [139, 140].
Sustained CTLA-4-Ig treatment increases survival, dampens autoantibody production and reduces kidney disease in BXSB and NZB/W mice, whereas a short-course therapy only delays disease [141–143]. Long-term treatment regimens are not likely to be used in clinical practice, thus short-term CTLA-4-Ig in combination with other immunosupressive reagents (anti-CD40L, CYC) has been evaluated in NZB/W mice, showing promising results [142, 144]. Importantly, the combination of CTLA-4-Ig with CYC reversed proteinuria in the majority of mice with advanced renal disease and precluded the need for continuous administration of CYC [144, 145].
Based on positive outcomes in mouse studies, the addition of abatacept to the CYC/AZA regimen has been evaluated in the ACCESS trial for the treatment of proliferative LN. Disappointingly, the randomized double-blinded study did not show any improvement [146]. As in the case of rituximab, the failure of abatacept trials has been partly attributed to clinical trial design, and post hoc analyses have shown potential benefit in patients with active arthritis and LN [140, 147]. The IFN signature in patients might also be considered, because IFNα renders NZB/W F1 mice relatively resistant to CYC/CTLA-4-Ig therapy [148]. Currently, the ALLURE trial is assessing the potential of abatacept for the treatment of advanced LN with CSs and MMF as background therapy [146].
IFNα- and TLR-targeted therapies
A prevalent IFN signature in the peripheral blood is present in > 80% lupus patients [8]. In BXSB and NZB/W models with a weak IFN signature, the blockade of the IFNα pathway either by mAb or by immunization with an IFNα kinoid ameliorated disease [20, 149]. A human IFNα kinoid has successfully reduced the IFNα signature in SLE patients [150]. The efficacy of anti-IFNα (rontalizumab, sifalimumab) or anti-IFNAR1 (anifrolumab) antibodies showed variable results depending on the patient IFN signature [151–153]. It has to be noted that the assessment of the IFN signature varies greatly between these studies, and a standardized panel of genes should be used, ideally involving IFNα-, IFNβ- and IFNγ-related modules, as recently described [8]. It also remains to be determined whether a high IFN signature in patients makes them more resistant to treatment as has been observed in NZB/W mice following IFNα exposure [148].
In one study, the IFN signature could be normalized transiently only with aggressive high-dose Mp pulse therapy, but not by oral CSs [154]. In NZB/W and TLR7.Tg7.6 mice, this unresponsiveness was attributable to TLR7/9-mediated chronic activation. A TLR7/9 antagonist (IRS954) enhanced the sensitivity to CSs and could be potentially useful as a CS-sparing drug [154]. Additional mouse studies support TLR7/8/9 targeting as an important therapeutic venue in SLE [7, 71]. Single TLR7 antagonist (IRS661), dual TLR7/9 (IRS954) or triple TLR7/8/9 antagonists (IMO-8400, IMO-9200 and CPG 52364) have all shown efficacy in MRL/lpr and NZB/W models [155–159]. Despite the promising results in murine models, TLR antagonist development for treating SLE patients has somewhat halted at preclinical or early clinical phases [160]. DV1179, a dual TLR7/9 antagonist developed by Dynavax, did not reduce IFN-regulated genes in SLE patients in a Phase 1b/2a study [161]. There might be several reasons for these failures, including TLR antagonists being useful only for a specific subpopulation of SLE patients [71].
Summary statement
Mouse models of lupus are an indispensable tool for the study of lupus pathogenesis, especially the pathways involved in loss of tolerance, autoantibody production and progression to end-organ disease, such as GN. In the last two decades, these studies have provided many new target molecules and triggered a revival in lupus drug development. The same models are also used in preclinical studies of new drugs to assess safety, dosage and efficacy. Despite the promising results obtained in mouse studies, the majority of the novel drugs, with the exception of belimumab and IFNα-blocking agents, failed in clinical trials. This has prompted the US Food and Drug Administration to release special guidelines for SLE drug development [2, 77]. Selection of the patient population that is most likely to respond to treatment, the number of recruited patients, defining the outcomes of treatment, establishing the appropriate duration of the trial and the contribution of existing treatment are the main challenges of SLE trial design [2, 77, 146]. Better study design with a larger number of patients and less stringent end-point measures has probably contributed to a successful outcome of belimumab and anti-IFNα trials [147]. The failure of certain therapeutics in clinical trials could also be attributed to lack of efficiency, simply because the biology is not completely understood. There are also obvious differences between the human and mouse immune system, with lymphocyte frequencies being the most obvious one [162]. Lastly, laboratory mice are housed in relatively germ-free conditions, whereas humans are constantly being exposed to pathogens that activate the immune system in various ways, including by TLR engagement and IFN signalling. The involvement of the microbiome in the development of autoimmunity is currently a hot topic of investigation [163].
To summarize, mouse models of lupus will continue to be indispensable in multiple aspects of SLE research. Novel drug targets should be assessed across multiple spontaneous SLE models and, ideally, also in an IFNα-dependent or accelerated model if the impact of the IFN signature is expected. Improved understanding of the biology and better clinical trial design will be likely to generate more success stories similar to belimumab and anti-IFN treatment.
Acknowledgements
T.C.’s and A.M.F.’s work is supported by core funding from A*STAR at the Singapore Immunology Network, Singapore.
Funding: No specific funding was received from any bodies in the public, commercial or not-for-profit sectors to carry out the work described in this manuscript.
Disclosure statement: The authors have declared no conflicts of interest.
References
- 1. Kaul A, Gordon C, Crow MK. et al. Systemic lupus erythematosus. Nat Rev Dis Primers 2016;2:16039. [DOI] [PubMed] [Google Scholar]
- 2. Rodríguez-Pintó I, Espinosa G, Cervera R.. The problems and pitfalls in systemic lupus erythematosus drug discovery. Expert Opin Drug Discovery 2016;11:525–7. [DOI] [PubMed] [Google Scholar]
- 3. Fairhurst AM, Wandstrat AE, Wakeland EK.. Systemic lupus erythematosus: multiple immunological phenotypes in a complex genetic disease. Adv Immunol 2006;92:1–69. [DOI] [PubMed] [Google Scholar]
- 4. Andrews BS, Eisenberg RA, Theofilopoulos AN. et al. Spontaneous murine lupus-like syndromes. Clinical and immunopathological manifestations in several strains. J Exp Med 1978;148:1198–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Du Y, Sanam S, Kate K, Mohan C.. Animal models of lupus and lupus nephritis. Curr Pharm Des 2015;21:2320–49. [DOI] [PubMed] [Google Scholar]
- 6. Schneider M, Mosca M, Pego-Reigosa JM. et al. Understanding remission in real-world lupus patients across five European countries. Lupus 2016;25:505–12. [DOI] [PubMed] [Google Scholar]
- 7. Celhar T, Magalhães R, Fairhurst AM.. TLR7 and TLR9 in SLE: when sensing self goes wrong. Immunol Res 2012;53:58–77. [DOI] [PubMed] [Google Scholar]
- 8. Banchereau R, Hong S, Cantarel B. et al. Personalized immunomonitoring uncovers molecular networks that stratify lupus patients. Cell 2016;165:551–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Crow MK. Type I interferon in the pathogenesis of lupus. J Immunol 2014;192:5459–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA, Nagata S.. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature 1992;356:314–7. [DOI] [PubMed] [Google Scholar]
- 11. Zhou T, Bluethmann H, Eldridge J, Berry K, Mountz JD.. Origin of CD4−CD8−B220+ T cells in MRL-lpr/lpr mice. Clues from a T cell receptor beta transgenic mouse. J Immunol 1993;150:3651–67. Epub 1993/04/15. [PubMed] [Google Scholar]
- 12. Jeltsch-David H, Muller S.. Neuropsychiatric systemic lupus erythematosus and cognitive dysfunction: the MRL-lpr mouse strain as a model. Autoimmun Rev 2014;13:963–73. [DOI] [PubMed] [Google Scholar]
- 13. Ghoreishi M, Dutz JP.. Murine models of cutaneous involvement in lupus erythematosus. Autoimmun Rev 2009;8:484–7. [DOI] [PubMed] [Google Scholar]
- 14. Lee YH, Song GG.. Associations between the FAS -670 A/G, -1377 G/A, and FASL -844 T/C polymorphisms and susceptibility to systemic lupus erythematosus: a meta-analysis. Clin Exp Rheumatol 2016;34:0634–40. [PubMed] [Google Scholar]
- 15. Moudi B, Salimi S, Farajian Mashhadi F, Sandoughi M, Zakeri Z.. Association of FAS and FAS ligand genes polymorphism and risk of systemic lupus erythematosus. Sci World J 2013;2013:176741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Tarbox JA, Keppel MP, Topcagic N. et al. Elevated double negative T cells in pediatric autoimmunity. J Clin Immunol 2014;34:594–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Crispín JC, Oukka M, Bayliss G. et al. Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol 2008;181:8761–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Lu Q, Shen N, Li XM, Chen SL.. Genomic view of IFN-α response in pre-autoimmune NZB/W and MRL/lpr mice. Genes Immun 2007;8:590–603. [DOI] [PubMed] [Google Scholar]
- 19. Hron JD, Peng SL.. Type I IFN protects against murine lupus. J Immunol 2004;173:2134–42. [DOI] [PubMed] [Google Scholar]
- 20. Baccala R, Gonzalez-Quintial R, Schreiber RD. et al. Anti-IFN-α/β receptor antibody treatment ameliorates disease in lupus-predisposed mice. J Immunol 2012;189:5976–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Virdis A, Tani C, Duranti E. et al. Early treatment with hydroxychloroquine prevents the development of endothelial dysfunction in a murine model of systemic lupus erythematosus. Arthritis Res Ther 2015;17:277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Rudofsky UH, Lawrence DA.. New Zealand mixed mice: a genetic systemic lupus erythematosus model for assessing environmental effects. Environ Health Perspect 1999;107:713–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Waters ST, Fu SM, Gaskin F. et al. NZM2328: a new mouse model of systemic lupus erythematosus with unique genetic susceptibility loci. Clin Immunol 2001;100:372–83. [DOI] [PubMed] [Google Scholar]
- 24. Morel L, Mohan C, Yu Y. et al. Functional dissection of systemic lupus erythematosus using congenic mouse strains. J Immunol 1997;158:6019–28. [PubMed] [Google Scholar]
- 25. Rozzo SJ, Allard JD, Choubey D. et al. Evidence for an interferon-inducible gene, Ifi202, in the susceptibility to systemic lupus. Immunity 2001;15:435–43. [DOI] [PubMed] [Google Scholar]
- 26. Bolland S, Yim YS, Tus K, Wakeland EK, Ravetch JV.. Genetic modifiers of systemic lupus erythematosus in FcγRIIB−/− mice. J Exp Med 2002;195:1167–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Pritchard NR, Cutler AJ, Uribe S. et al. Autoimmune-prone mice share a promoter haplotype associated with reduced expression and function of the Fc receptor FcγRII. Curr Biol 2000;10:227–30. [DOI] [PubMed] [Google Scholar]
- 28. Wandstrat AE, Nguyen C, Limaye N. et al. Association of extensive polymorphisms in the SLAM/CD2 gene cluster with murine lupus. Immunity 2004;21:769–80. [DOI] [PubMed] [Google Scholar]
- 29. Kumar KR, Li L, Yan M. et al. Regulation of B cell tolerance by the lupus susceptibility gene Ly108. Science 2006;312:1665–9. [DOI] [PubMed] [Google Scholar]
- 30. Jørgensen TN, Alfaro J, Enriquez HL. et al. Development of murine lupus involves the combined genetic contribution of the SLAM and FcγR intervals within the Nba2 autoimmune susceptibility locus. J Immunol 2010;184:775–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Tsao BP, Cantor RM, Kalunian KC. et al. Evidence for linkage of a candidate chromosome 1 region to human systemic lupus erythematosus. J Clin Invest 1997;99:725–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Moser KL, Neas BR, Salmon JE. et al. Genome scan of human systemic lupus erythematosus: evidence for linkage on chromosome 1q in African-American pedigrees. Proc Natl Acad Sci USA 1998;95:14869–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Shai R, Quismorio FP Jr, Li L. et al. Genome-wide screen for systemic lupus erythematosus susceptibility genes in multiplex families. Human Mol Genetics 1999;8:639–44. [DOI] [PubMed] [Google Scholar]
- 34. Boackle SA, Holers VM, Chen X. et al. Cr2, a candidate gene in the murine Sle1c lupus susceptibility locus, encodes a dysfunctional protein. Immunity 2001;15:775–85. [DOI] [PubMed] [Google Scholar]
- 35. Zhao J, Giles BM, Taylor RL. et al. Preferential association of a functional variant in complement receptor 2 with antibodies to double-stranded DNA. Ann Rheum Dis 2016;75:242–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Salmon JE, Millard S, Schachter LA. et al. FcγRIIA alleles are heritable risk factors for lupus nephritis in African Americans. J Clin Invest 1996;97:1348–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Wu J, Edberg JC, Redecha PB. et al. A novel polymorphism of FcγRIIIa (CD16) alters receptor function and predisposes to autoimmune disease. J Clin Invest 1997;100:1059–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Tsang ASMW, Nagelkerke SQ, Bultink IE. et al. Fc-gamma receptor polymorphisms differentially influence susceptibility to systemic lupus erythematosus and lupus nephritis. Rheumatology 2016;55:939–48. [DOI] [PubMed] [Google Scholar]
- 39. Tsao BP, Cantor RM, Grossman JM. et al. PARP alleles within the linked chromosomal region are associated with systemic lupus erythematosus. J Clin Invest 1999;103:1135–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Furukawa H, Kawasaki A, Oka S. et al. Association of a single nucleotide polymorphism in the SH2D1A intronic region with systemic lupus erythematosus. Lupus 2013;22:497–503. [DOI] [PubMed] [Google Scholar]
- 41. Russell AI, Cunninghame Graham DS, Shepherd C. et al. Polymorphism at the C-reactive protein locus influences gene expression and predisposes to systemic lupus erythematosus. Hum Mol Genet 2004;13:137–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Wong EB, Soni C, Chan AY. et al. B cell-intrinsic CD84 and Ly108 maintain germinal center B cell tolerance. J Immunol 2015;194:4130–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Ge Y, Jiang C, Sung SS. et al. Cgnz1 allele confers kidney resistance to damage preventing progression of immune complex-mediated acute lupus glomerulonephritis. J Exp Med 2013;210:2387–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Mohan C, Yu Y, Morel L, Yang P, Wakeland EK.. Genetic dissection of Sle pathogenesis: Sle3 on murine chromosome 7 impacts T cell activation, differentiation, and cell death. J Immunol 1999;162:6492–502. [PubMed] [Google Scholar]
- 45. Liu K, Li QZ, Yu Y. et al. Sle3 and Sle5 can independently couple with Sle1 to mediate severe lupus nephritis. Genes Immun 2007;8:634–45. [DOI] [PubMed] [Google Scholar]
- 46. Liu K, Li QZ, Delgado-Vega AM. et al. Kallikrein genes are associated with lupus and glomerular basement membrane-specific antibody-induced nephritis in mice and humans. J Clin Invest 2009;119:911–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Zeumer L, Sang A, Niu H, Morel L.. Murine lupus susceptibility locus Sle2 activates DNA-reactive B cells through two sub-loci with distinct phenotypes. Genes Immun 2011;12:199–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Waters ST, McDuffie M, Bagavant H. et al. Breaking tolerance to double stranded DNA, nucleosome, and other nuclear antigens is not required for the pathogenesis of lupus glomerulonephritis. J Exp Med 2004;199:255–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Sriram U, Varghese L, Bennett HL. et al. Myeloid dendritic cells from B6.NZM Sle1/Sle2/Sle3 lupus-prone mice express an IFN signature that precedes disease onset. J Immunol 2012;189:80–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Agrawal H, Jacob N, Carreras E. et al. Deficiency of type I IFN receptor in lupus-prone New Zealand mixed 2328 mice decreases dendritic cell numbers and activation and protects from disease. J Immunol 2009;183:6021–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Jørgensen TN, Roper E, Thurman JM, Marrack P, Kotzin BL.. Type I interferon signaling is involved in the spontaneous development of lupus-like disease in B6.Nba2 and (B6.Nba2 × NZW)F1 mice. Genes Immun 2007;8:653–62. [DOI] [PubMed] [Google Scholar]
- 52. Subramanian S, Tus K, Li QZ. et al. A Tlr7 translocation accelerates systemic autoimmunity in murine lupus. Proc Natl Acad Sci USA 2006;103:9970–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Pisitkun P, Deane JA, Difilippantonio MJ. et al. Autoreactive B cell responses to RNA-related antigens due to TLR7 gene duplication. Science 2006;312:1669–72. [DOI] [PubMed] [Google Scholar]
- 54. Deane JA, Pisitkun P, Barrett RS. et al. Control of toll-like receptor 7 expression is essential to restrict autoimmunity and dendritic cell proliferation. Immunity 2007;27:801–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Fairhurst AM, Hwang SH, Wang A. et al. Yaa autoimmune phenotypes are conferred by overexpression of TLR7. Eur J Immunol 2008;38:1971–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Santiago-Raber ML, Kikuchi S, Borel P. et al. Evidence for genes in addition to Tlr7 in the Yaa translocation linked with acceleration of systemic lupus erythematosus. J Immunol 2008;181:1556–62. [DOI] [PubMed] [Google Scholar]
- 57. Hudgins CC, Steinberg RT, Klinman DM, Reeves MJ, Steinberg AD.. Studies of consomic mice bearing the Y chromosome of the BXSB mouse. J Immunol 1985;134:3849–54. [PubMed] [Google Scholar]
- 58. Merino R, Shibata T, De Kossodo S, Izui S.. Differential effect of the autoimmune Yaa and lpr genes on the acceleration of lupus-like syndrome in MRL/MpJ mice. Eur J Immunol 1989;19:2131–7. [DOI] [PubMed] [Google Scholar]
- 59. Hwang SH, Lee H, Yamamoto M. et al. B cell TLR7 expression drives anti-RNA autoantibody production and exacerbates disease in systemic lupus erythematosus-prone mice. J Immunol 2012;189:5786–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Izui S, Higaki M, Morrow D, Merino R.. The Y chromosome from autoimmune BXSB/MpJ mice induces a lupus-like syndrome in (NZW × C57BL/6)F1 male mice, but not in C57BL/6 male mice. Eur J Immunol 1988;18:911–5. [DOI] [PubMed] [Google Scholar]
- 61. Hogarth MB, Slingsby JH, Allen PJ. et al. Multiple lupus susceptibility loci map to chromosome 1 in BXSB mice. J Immunol 1998;161:2753–61. [PubMed] [Google Scholar]
- 62. Haywood ME, Hogarth MB, Slingsby JH. et al. Identification of intervals on chromosomes 1, 3, and 13 linked to the development of lupus in BXSB mice. Arthritis Rheum 2000;43:349–55. [DOI] [PubMed] [Google Scholar]
- 63. Boswell J, Schur PH.. Monocyte function in systemic lupus erythematosus. Clin Immunol Immunopathol 1989;52:271–8. [DOI] [PubMed] [Google Scholar]
- 64. Mukherjee R, Kanti Barman P, Kumar Thatoi P. et al. Non-classical monocytes display inflammatory features: validation in sepsis and systemic lupus erythematous. Sci Rep 2015;5:13886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Haywood ME, Rogers NJ, Rose SJ. et al. Dissection of BXSB lupus phenotype using mice congenic for chromosome 1 demonstrates that separate intervals direct different aspects of disease. J Immunol 2004;173:4277–85. [DOI] [PubMed] [Google Scholar]
- 66. Dillon SP, Kurien BT, Li S. et al. Sex chromosome aneuploidies among men with systemic lupus erythematosus. J Autoimmun 2012;38:J129–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Lee YH, Choi SJ, Ji JD, Song GG.. Association between toll-like receptor polymorphisms and systemic lupus erythematosus: a meta-analysis update. Lupus 2016;25:593–601. [DOI] [PubMed] [Google Scholar]
- 68. García-Ortiz H, Velázquez-Cruz R, Espinosa-Rosales F. et al. Association of TLR7 copy number variation with susceptibility to childhood-onset systemic lupus erythematosus in Mexican population. Ann Rheum Dis 2010;69:1861–5. [DOI] [PubMed] [Google Scholar]
- 69. Niewold TB, Kelly JA, Flesch MH. et al. Association of the IRF5 risk haplotype with high serum interferon-α activity in systemic lupus erythematosus patients. Arthritis Rheum 2008;58:2481–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Kelly JA, Kelley JM, Kaufman KM. et al. Interferon regulatory factor-5 is genetically associated with systemic lupus erythematosus in African Americans. Genes Immun 2008;9:187–94. [DOI] [PubMed] [Google Scholar]
- 71. Celhar T, Fairhurst AM.. Toll-like receptors in systemic lupus erythematosus: potential for personalized treatment. Front Pharmacol 2014;5:265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Garcia-Romo GS, Caielli S, Vega B. et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Trans Med 2011;3:73ra20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Lande R, Ganguly D, Facchinetti V. et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA–peptide complexes in systemic lupus erythematosus. Sci Trans Med 2011;3:73ra19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Rowland SL, Riggs JM, Gilfillan S. et al. Early, transient depletion of plasmacytoid dendritic cells ameliorates autoimmunity in a lupus model. J Exp Med 2014;211:1977–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Reeves WH, Lee PY, Weinstein JS, Satoh M, Lu L.. Induction of autoimmunity by pristane and other naturally occurring hydrocarbons. Trends Immunol 2009;30:455–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Liu Z, Davidson A.. IFNα inducible models of murine SLE. Front Immunol 2013;4:306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wallace DJ. The evolution of drug discovery in systemic lupus erythematosus. Nat Rev Rheumatol 2015;11:616–20. [DOI] [PubMed] [Google Scholar]
- 78. Muangchan C, van Vollenhoven RF, Bernatsky SR. et al. Treatment algorithms in systemic lupus erythematosus. Arthritis Care Res 2015;67:1237–45. [DOI] [PubMed] [Google Scholar]
- 79. Emadi A, Jones RJ, Brodsky RA.. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol 2009;6:638–47. [DOI] [PubMed] [Google Scholar]
- 80. Russell PJ, Hicks JD.. Cyclophosphamide treatment of renal disease in (NZB × NZW) F1 hybrid mice. Lancet 1968;1:440–1. Epub 1968/03/02. [DOI] [PubMed] [Google Scholar]
- 81. Casey TP. Immunosuppression by cyclophosphamide in NZB X NZW mice with lupus nephritis. Blood 1968;32:436–44. [PubMed] [Google Scholar]
- 82. Walker SE, Bole GG Jr.. Selective suppression of autoantibody responses in NZB/NZW mice treated with long-term cyclophosphamide. Arthritis Rheum 1975;18:265–72. [DOI] [PubMed] [Google Scholar]
- 83. Austin HA 3rd, Patel AD, Cadena CA, Boumpas DT, Balow JE.. Ongoing immunologic activity after short courses of pulse cyclophosphamide in the NZB/W murine model of systemic lupus erythematosus. J Rheumatol 1997;24:61–8. [PubMed] [Google Scholar]
- 84. Mathian A, Gallegos M, Pascual V, Banchereau J, Koutouzov S.. Interferon-α induces unabated production of short-lived plasma cells in pre-autoimmune lupus-prone (NZB×NZW)F1 mice but not in BALB/c mice. Eur J Immunol 2011;41:863–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Smith HR, Chused TM, Steinberg AD.. Cyclophosphamide-induced changes in the MRL-lpr/lpr mouse: effects upon cellular composition, immune function, and disease. Clin Immunol Immunopathol 1984;30:51–61. [DOI] [PubMed] [Google Scholar]
- 86. Shiraki M, Fujiwara M, Tomura S.. Long term administration of cyclophosphamide in MRL/1 mice. I. The effects on the development of immunological abnormalities and lupus nephritis. Clin Exp Immunol 1984;55:333–9. [PMC free article] [PubMed] [Google Scholar]
- 87. Hoyer BF, Moser K, Hauser AE. et al. Short-lived plasmablasts and long-lived plasma cells contribute to chronic humoral autoimmunity in NZB/W mice. J Exp Med 2004;199:1577–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Cassese G, Lindenau S, de Boer B. et al. Inflamed kidneys of NZB/W mice are a major site for the homeostasis of plasma cells. Eur J Immunol 2001;31:2726–32. [DOI] [PubMed] [Google Scholar]
- 89. Taddeo A, Khodadadi L, Voigt C. et al. Long-lived plasma cells are early and constantly generated in New Zealand Black/New Zealand White F1 mice and their therapeutic depletion requires a combined targeting of autoreactive plasma cells and their precursors. Arthritis Res Therapy 2015;17:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Mumtaz IM, Hoyer BF, Panne D. et al. Bone marrow of NZB/W mice is the major site for plasma cells resistant to dexamethasone and cyclophosphamide: implications for the treatment of autoimmunity. J Autoimmun 2012;39:180–8. [DOI] [PubMed] [Google Scholar]
- 91. Zickert A, Sundelin B, Svenungsson E, Gunnarsson I.. Role of early repeated renal biopsies in lupus nephritis. Lupus Sci Med 2014;1:e000018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Hahn BH, Knotts L, Ng M, Hamilton TR.. Influence of cyclophosphamide and other immunosuppressive drugs on immune disorders and neoplasia in NZB/NZW mice. Arthritis Rheum 1975;18:145–52. [DOI] [PubMed] [Google Scholar]
- 93. Walker SE, Anver MR.. Accelerated appearance of neoplasms in female NZB/NZW mice treated with high-dose cyclophosphamide. Arthritis Rheum 1979;22:1338–43. [DOI] [PubMed] [Google Scholar]
- 94. Steinberg AD, Gelfand MC, Hardin JA, Lowenthal DT.. Therapeutic studies in NZB/W mice. III. Relationship between renal status and efficacy of immunosuppressive drug therapy. Arthritis Rheum 1975;18:9–14. [DOI] [PubMed] [Google Scholar]
- 95. Steinberg AD, Steinberg SC.. Long-term preservation of renal function in patients with lupus nephritis receiving treatment that includes cyclophosphamide versus those treated with prednisone only. Arthritis Rheum 1991;34:945–50. [DOI] [PubMed] [Google Scholar]
- 96. Austin HA 3rd, Klippel JH, Balow JE. et al. Therapy of lupus nephritis. Controlled trial of prednisone and cytotoxic drugs. New Engl J Med 1986;314:614–9. [DOI] [PubMed] [Google Scholar]
- 97. Boumpas DT, Chrousos GP, Wilder RL, Cupps TR, Balow JE.. Glucocorticoid therapy for immune-mediated diseases: basic and clinical correlates. Ann Int Med 1993;119:1198–208. [DOI] [PubMed] [Google Scholar]
- 98. Cavallo T, Graves K, Granholm NA.. Murine lupus nephritis. Effects of glucocorticoid on circulating and tissue-bound immunoreactants. Lab Invest 1983;49:476–81. [PubMed] [Google Scholar]
- 99. Cavallo T, Graves K, Granholm NA.. Murine lupus nephritis. Effects of glucocorticoid on glomerular permeability. Lab Invest 1984;50:378–84. [PubMed] [Google Scholar]
- 100. De Bosscher K, Vanden Berghe W, Haegeman G.. The interplay between the glucocorticoid receptor and nuclear factor-κB or activator protein-1: molecular mechanisms for gene repression. Endocr Rev 2003;24:488–522. [DOI] [PubMed] [Google Scholar]
- 101. Kalergis AM, Iruretagoyena MI, Barrientos MJ. et al. Modulation of nuclear factor-κB activity can influence the susceptibility to systemic lupus erythematosus. Immunology 2009;128:e306–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. You Y, Qin Y, Lin X. et al. Methylprednisolone attenuates lipopolysaccharide-induced Fractalkine expression in kidney of lupus-prone MRL/lpr mice through the NF-kappaB pathway. BMC Nephrol 2015;16:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Teramoto K, Negoro N, Kitamoto K. et al. Microarray analysis of glomerular gene expression in murine lupus nephritis. J Pharmacol Sci 2008;106:56–67. [DOI] [PubMed] [Google Scholar]
- 104. Pahl HL. Activators and target genes of Rel/NF-κB transcription factors. Oncogene 1999;18:6853–66. [DOI] [PubMed] [Google Scholar]
- 105. Nakamura T, Ebihara I, Tomino Y, Koide H.. Glucocorticoid ameliorates altered gene expression of extracellular matrix components in kidneys of New Zealand black/white F1 mice. Clin Sci 1992;83:701–9. [DOI] [PubMed] [Google Scholar]
- 106. Corna D, Morigi M, Facchinetti D. et al. Mycophenolate mofetil limits renal damage and prolongs life in murine lupus autoimmune disease. Kidney Int 1997;51:1583–9. [DOI] [PubMed] [Google Scholar]
- 107. McMurray RW, Elbourne KB, Lagoo A, Lal S.. Mycophenolate mofetil suppresses autoimmunity and mortality in the female NZB × NZW F1 mouse model of systemic lupus erythematosus. J Rheumatol 1998;25:2364–70. [PubMed] [Google Scholar]
- 108. Ramos MA, Piñera C, Setién MA. et al. Modulation of autoantibody production by mycophenolate mofetil: effects on the development of SLE in (NZB×NZW)F1 mice. Nephrol Dial Transplant 2003;18:878–83. [DOI] [PubMed] [Google Scholar]
- 109. Van Bruggen MC, Walgreen B, Rijke TP, Berden JH.. Attenuation of murine lupus nephritis by mycophenolate mofetil. J Am Soc Nephrol 1998;9:1407–15. [DOI] [PubMed] [Google Scholar]
- 110. Jonsson CA, Svensson L, Carlsten H.. Beneficial effect of the inosine monophosphate dehydrogenase inhibitor mycophenolate mofetil on survival and severity of glomerulonephritis in systemic lupus erythematosus (SLE)-prone MRLlpr/lpr mice. Clin Exp Immunol 1999;116:534–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Haselmayer P, Camps M, Muzerelle M. et al. Characterization of novel PI3Kδ inhibitors as potential therapeutics for SLE and lupus nephritis in pre-clinical studies. Front Immunol 2014;5:233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Yu CC, Yang CW, Wu MS. et al. Mycophenolate mofetil reduces renal cortical inducible nitric oxide synthase mRNA expression and diminishes glomerulosclerosis in MRL/lpr mice. J Lab Clin Med 2001;138:69–77. [DOI] [PubMed] [Google Scholar]
- 113. Lui SL, Tsang R, Wong D. et al. Effect of mycophenolate mofetil on severity of nephritis and nitric oxide production in lupus-prone MRL/lpr mice. Lupus 2002;11:411–8. [DOI] [PubMed] [Google Scholar]
- 114. Jonsson CA, Erlandsson M, Svensson L, Mölne J, Carlsten H.. Mycophenolate mofetil ameliorates perivascular T lymphocyte inflammation and reduces the double-negative T cell population in SLE-prone MRLlpr/lpr mice. Cell Immunol 1999;197:136–44. [DOI] [PubMed] [Google Scholar]
- 115. Cheng CC, Lee YF, Lan JL. et al. Mycophenolate mofetil alleviates lupus nephritis through urokinase receptor signaling in a mice model. Lupus 2013;22:554–61. [DOI] [PubMed] [Google Scholar]
- 116. Yung S, Zhang Q, Zhang CZ. et al. Anti-DNA antibody induction of protein kinase C phosphorylation and fibronectin synthesis in human and murine lupus and the effect of mycophenolic acid. Arthritis Rheum 2009;60:2071–82. [DOI] [PubMed] [Google Scholar]
- 117. Lee SJ, Silverman E, Bargman JM.. The role of antimalarial agents in the treatment of SLE and lupus nephritis. Nat Rev Nephrol 2011;7:718–29. [DOI] [PubMed] [Google Scholar]
- 118. Wallace DJ, Gudsoorkar VS, Weisman MH, Venuturupalli SR.. New insights into mechanisms of therapeutic effects of antimalarial agents in SLE. Nat Rev Rheumatol 2012;8:522–33. [DOI] [PubMed] [Google Scholar]
- 119. Gómez-Guzmán M, Jiménez R, Romero M. et al. Chronic hydroxychloroquine improves endothelial dysfunction and protects kidney in a mouse model of systemic lupus erythematosus. Hypertension 2014;64:330–7. [DOI] [PubMed] [Google Scholar]
- 120. Shimomatsu T, Kanazawa N, Mikita N. et al. The effect of hydroxychloroquine on lupus erythematosus-like skin lesions in MRL/lpr mice. Mod Rheumatol 2016;26:744–8. [DOI] [PubMed] [Google Scholar]
- 121. Stohl W, Hilbert DM.. The discovery and development of belimumab: the anti-BLyS–lupus connection. Nat Biotechnol 2012;30:69–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Vincent FB, Morand EF, Schneider P, Mackay F.. The BAFF/APRIL system in SLE pathogenesis. Nat Rev Rheumatol 2014;10:365–73. [DOI] [PubMed] [Google Scholar]
- 123. Moore PA, Belvedere O, Orr A. et al. BLyS: member of the tumor necrosis factor family and B lymphocyte stimulator. Science 1999;285:260–3. [DOI] [PubMed] [Google Scholar]
- 124. Mackay F, Woodcock SA, Lawton P. et al. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med 1999;190:1697–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Stohl W, Xu D, Kim KS. et al. BAFF overexpression and accelerated glomerular disease in mice with an incomplete genetic predisposition to systemic lupus erythematosus. Arthritis Rheumeum 2005;52:2080–91. [DOI] [PubMed] [Google Scholar]
- 126. Ramanujam M, Wang X, Huang W. et al. Similarities and differences between selective and nonselective BAFF blockade in murine SLE. J Clin Invest 2006;116:724–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Ding H, Wang L, Wu X. et al. Blockade of B-cell-activating factor suppresses lupus-like syndrome in autoimmune BXSB mice. J Cell Mol Med 2010;14:1717–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Gross JA, Johnston J, Mudri S. et al. TACI and BCMA are receptors for a TNF homologue implicated in B-cell autoimmune disease. Nature 2000;404:995–9. [DOI] [PubMed] [Google Scholar]
- 129. Jacob N, Guo S, Mathian A. et al. B Cell and BAFF dependence of IFN-α-exaggerated disease in systemic lupus erythematosus-prone NZM 2328 mice. J Immunol 2011;186:4984–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Thien M, Phan TG, Gardam S. et al. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity 2004;20:785–98. [DOI] [PubMed] [Google Scholar]
- 131. Figgett WA, Deliyanti D, Fairfax KA. et al. Deleting the BAFF receptor TACI protects against systemic lupus erythematosus without extensive reduction of B cell numbers. J Autoimmun 2015;61:9–16. [DOI] [PubMed] [Google Scholar]
- 132. Kamal A, Khamashta M.. The efficacy of novel B cell biologics as the future of SLE treatment: a review. Autoimmun Rev 2014;13:1094–101. [DOI] [PubMed] [Google Scholar]
- 133. Ahuja A, Shupe J, Dunn R. et al. Depletion of B cells in murine lupus: efficacy and resistance. J Immunol 2007;179:3351–61. [DOI] [PubMed] [Google Scholar]
- 134. Ahuja A, Teichmann LL, Wang H. et al. An acquired defect in IgG-dependent phagocytosis explains the impairment in antibody-mediated cellular depletion in lupus. J Immunol 2011;187:3888–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Bekar KW, Owen T, Dunn R. et al. Prolonged effects of short-term anti-CD20 B cell depletion therapy in murine systemic lupus erythematosus. Arthritis Rheum 2010;62:2443–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Wang W, Rangel-Moreno J, Owen T. et al. Long-term B cell depletion in murine lupus eliminates autoantibody-secreting cells and is associated with alterations in the kidney plasma cell niche. J Immunol 2014;192:3011–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Lin W, Seshasayee D, Lee WP. et al. Dual B cell immunotherapy is superior to individual anti-CD20 depletion or BAFF blockade in murine models of spontaneous or accelerated lupus. Arthritis Rheumatol 2015;67:215–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Cobo-Ibáñez T, Loza-Santamaría E, Pego-Reigosa JM. et al. Efficacy and safety of rituximab in the treatment of non-renal systemic lupus erythematosus: a systematic review. Semin Arthritis Rheum 2014;44:175–85. [DOI] [PubMed] [Google Scholar]
- 139. Romo-Tena J, Gómez-Martín D, Alcocer-Varela J.. CTLA-4 and autoimmunity: new insights into the dual regulator of tolerance. Autoimmun Rev 2013;12:1171–6. [DOI] [PubMed] [Google Scholar]
- 140. Mok CC. Abatacept for systemic lupus erythematosus: the outlook. Expert Opin Biol Ther 2012;12:1559–61. [DOI] [PubMed] [Google Scholar]
- 141. Chu EB, Hobbs MV, Wilson CB. et al. Intervention of CD4+ cell subset shifts and autoimmunity in the BXSB mouse by murine CTLA4Ig. J Immunol 1996;156:1262–8. [PubMed] [Google Scholar]
- 142. Daikh DI, Finck BK, Linsley PS, Hollenbaugh D, Wofsy D.. Long-term inhibition of murine lupus by brief simultaneous blockade of the B7/CD28 and CD40/gp39 costimulation pathways. J Immunol 1997;159:3104–8. [PubMed] [Google Scholar]
- 143. Mihara M, Tan I, Chuzhin Y. et al. CTLA4Ig inhibits T cell-dependent B-cell maturation in murine systemic lupus erythematosus. J Clin Invest 2000;106:91–101. Epub 2000/07/06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Daikh DI, Wofsy D.. Cutting edge: reversal of murine lupus nephritis with CTLA4Ig and cyclophosphamide. J Immunol 2001;166:2913–6. [DOI] [PubMed] [Google Scholar]
- 145. Cunnane G, Chan OT, Cassafer G. et al. Prevention of renal damage in murine lupus nephritis by CTLA-4Ig and cyclophosphamide. Arthritis Rheum 2004;50:1539–48. [DOI] [PubMed] [Google Scholar]
- 146. Corapi KM, Dooley MA, Pendergraft WF 3rd.. Comparison and evaluation of lupus nephritis response criteria in lupus activity indices and clinical trials. Arthritis Res Ther 2015;17:110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Gatto M, Saccon F, Zen M. et al. Success and failure of biological treatment in systemic lupus erythematosus: a critical analysis. J Autoimmun 2016;74:94–105. [DOI] [PubMed] [Google Scholar]
- 148. Liu Z, Bethunaickan R, Huang W. et al. IFN-α confers resistance of systemic lupus erythematosus nephritis to therapy in NZB/W F1 mice. J Immunol 2011;187:1506–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Zagury D, Le Buanec H, Mathian A. et al. IFNα kinoid vaccine-induced neutralizing antibodies prevent clinical manifestations in a lupus flare murine model. Proc Natl Acad Sci USA 2009;106:5294–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Lauwerys BR, Hachulla E, Spertini F. et al. Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon α-kinoid. Arthritis Rheum 2013;65:447–56. [DOI] [PubMed] [Google Scholar]
- 151. Kalunian KC, Merrill JT, Maciuca R. et al. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-α) in patients with systemic lupus erythematosus (ROSE). Ann Rheum Dis 2016;75:196–202. [DOI] [PubMed] [Google Scholar]
- 152. Khamashta M, Merrill JT, Werth VP. et al. Sifalimumab, an anti-interferon-α monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis 2016;75:1909–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Furie RMJ, Werth V, Khamashta M. et al. Anifrolumab, an anti-interferon alpha receptor monoclonal antibody, in moderate to severe systemic lupus erythematosus (SLE) [abstract]. Arthritis Rheumatol 2015;67:2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Guiducci C, Gong M, Xu Z. et al. TLR recognition of self nucleic acids hampers glucocorticoid activity in lupus. Nature 2010;465:937–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Pawar RD, Ramanjaneyulu A, Kulkarni OP. et al. Inhibition of Toll-like receptor-7 (TLR-7) or TLR-7 plus TLR-9 attenuates glomerulonephritis and lung injury in experimental lupus. J Am Soc Nephrol 2007;18:1721–31. [DOI] [PubMed] [Google Scholar]
- 156. Barrat FJ, Meeker T, Chan JH, Guiducci C, Coffman RL.. Treatment of lupus-prone mice with a dual inhibitor of TLR7 and TLR9 leads to reduction of autoantibody production and amelioration of disease symptoms. Eur J Immunol 2007;37:3582–6. [DOI] [PubMed] [Google Scholar]
- 157. Zhu FG, Jiang W, Bhagat L. et al. A novel antagonist of Toll-like receptors 7, 8 and 9 suppresses lupus disease-associated parameters in NZBW/F1 mice. Autoimmunity 2013;46:419–28. [DOI] [PubMed] [Google Scholar]
- 158. Lipford G, Forsbach A, Zepp C. et al. Selective Toll-like Receptor 7/8/9 Antagonists for the Oral Treatment of Autoimmune Diseases. Abstract number 1596. American College of Rheumatology, 2007 Annual Scientific Meeting, November 10-11, 2007 https://acr.confex.com/acr/2007/webprogram/Paper8044.html
- 159. Bhagat L, Wang D, Jiang W, IMO-9200, a novel clinical-stage TLR antagonist for the treatment of autoimmune diseases, inhibits TLR-mediated immune responses and shows activity in animal modes. 10th Annual Meeting of the Oligonucleotide Therapeutics Society. San Diego, CA, October 12-15, 2014.
- 160. Junt T, Barchet W.. Translating nucleic acid-sensing pathways into therapies. Nat Rev Immunol 2015;15:529–44. [DOI] [PubMed] [Google Scholar]
- 161. Ostrach M. Dynavax regains full rights to investigational TLR 7/9 inhibitor DV1179 following expiration of collaboration with GSK. 2014. http://investors.dynavax.com/releasedetail.cfm?releaseid=885172 (29 June 2016, date last accessed).
- 162. Mestas J, Hughes CC.. Of mice and not men: differences between mouse and human immunology. J Immunol 2004;172:2731–8. [DOI] [PubMed] [Google Scholar]
- 163. Shamriz O, Mizrahi H, Werbner M. et al. Microbiota at the crossroads of autoimmunity. Autoimmun Rev 2016;15:859–69. [DOI] [PubMed] [Google Scholar]