Defective DNA repair and chromatin organization in patients with ...

2 downloads 113 Views 4MB Size Report
Efficiency of the DNA repair and polymorphisms of the XRCC1, XRCC3 and XRCC4 DNA repair genes in systemic lupus erythematosus. Lupus. 2008 ...
Souliotis et al. Arthritis Research & Therapy (2016) 18:182 DOI 10.1186/s13075-016-1081-3

RESEARCH ARTICLE

Open Access

Defective DNA repair and chromatin organization in patients with quiescent systemic lupus erythematosus Vassilis L. Souliotis1,2*, Konstantinos Vougas3, Vassilis G. Gorgoulis3,4 and Petros P. Sfikakis2

Abstract Background: Excessive autoantibody production characterizing systemic lupus erythematosus (SLE) occurs irrespective of the disease’s clinical status and is linked to increased lymphocyte apoptosis. Herein, we tested the hypothesis that defective DNA damage repair contributes to increased apoptosis in SLE. Methods: We evaluated nucleotide excision repair at the N-ras locus, DNA double-strand breaks repair and apoptosis rates in peripheral blood mononuclear cells from anti-dsDNA autoantibody-positive patients (six with quiescent disease and six with proliferative nephritis) and matched healthy controls following ex vivo treatment with melphalan. Chromatin organization and expression levels of DNA repair- and apoptosis-associated genes were also studied in quiescent SLE. Results: Defective nucleotide excision repair and DNA double-strand breaks repair were found in SLE, with lupus nephritis patients showing higher DNA damage levels than those with quiescent disease. Melphalan-induced apoptosis rates were higher in SLE than control cells and correlated inversely with DNA repair efficiency. Chromatin at the N-ras locus was more condensed in SLE than controls, while treatment with the histone deacetylase inhibitor vorinostat resulted in hyperacetylation of histone H4, chromatin decondensation, amelioration of DNA repair efficiency and decreased apoptosis. Accordingly, genes involved in DNA damage repair and signaling pathways, such as DDB1, ERCC2, XPA, XPC, MRE11A, RAD50, PARP1, MLH1, MLH3, and ATM were significantly underexpressed in SLE versus controls, whereas PPP1R15A, BARD1 and BBC3 genes implicated in apoptosis were significantly overexpressed. Conclusions: Epigenetically regulated functional abnormalities of DNA repair machinery occur in SLE, regardless of clinical disease activity, and may promote lymphocyte apoptosis. Approaches to correct these abnormalities may be of therapeutic value in SLE. Keywords: Systemic lupus erythematosus, Nucleotide excision repair, DNA double-strand breaks repair, Chromatin organization, Apoptosis, Histone deacetylase inhibitor

Background The prototypic systemic autoimmune disease, systemic lupus erythematosus (SLE), is characterized by loss of immunological tolerance to self-nuclear antigens and abnormal T- and B-cell responses, which are attributed to genetic and epigenetic causes [1]. Excessive autoantigen * Correspondence: [email protected] 1 Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece 2 Joint Rheumatology Program and First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece Full list of author information is available at the end of the article

presentation and autoantibody production is considered a central pathogenetic mechanism of SLE [2]. Significant diversity in organ involvement, disease activity, severity and responsiveness to treatment are common [3]. Of note, the production of myriads of autoantibodies in human SLE may precede the onset of clinical disease by many years and is facilitated by accumulation of dead cells due to dysregulated apoptosis [1, 4]. Every day, each of our cells is faced with at least 104 DNA lesions and their efficient repair is a mandatory biological process to preserve cell survival and function [5]. The genomic integrity is maintained by a network of

© 2016 The Author(s). Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

DNA damage response and repair pathways, a comprehensive signaling process that determines the cell's ability to repair DNA damage or to undergo mutation(s) or apoptosis [6]. The wide diversity of DNA lesion types necessitates multiple, largely distinct DNA repair mechanisms. For example, nucleotide excision repair executes the repair of DNA lesions induced by UV light, polycyclic aromatic hydrocarbons, heavy metals such as platinum and cadmium, and by other sources that form bulky DNA lesions that distort the DNA helix [7]. On the other hand, for DNA double-strand breaks, which are introduced by exogenous agents such as ionizing radiation and therapeutic drugs as well as by cellular processes such as V(D)J recombination, class switch recombination, stalled replication forks and reactions that generate reactive oxygen species [8], two principal repair mechanisms are used. The first is the homologous recombination, an error-free repair pathway that uses DNA homology to direct DNA repair, and the second is the nonhomologous end joining, a straightforward religation of broken DNA ends without any requirement for a template [5]. Defective nucleotide excision repair has been indeed reported in SLE [9–11], whereas the majority of B lymphoblastoid cell lines derived from children with SLE gave evidence of inefficient or delayed kinetics of DNA double-strand breaks repair [12]. Our recent study has shown that circulating mononuclear cells from patients with a severe SLE complication, such as lupus nephritis, are characterized by higher intrinsic DNA double-strand breaks accumulation compared to healthy controls, possibly due to increased susceptibility of these cells to develop these DNA lesions and/or an impaired repair [13]. Herein, we considered that increased apoptosis in SLE could be due, at least in part, to defective mechanisms of DNA damage repair, thus providing an additional putative pathogenetic mechanism in systemic autoimmunity. Indeed, we found that nucleotide excision repair and DNA double-strand breaks repair mechanisms are defective in patients with either clinically quiescent disease or proliferative lupus nephritis, and strongly correlate with increased apoptosis. Notably, treatment of lupus mononuclear cells with the histone deacetylase inhibitor vorinostat resulted in amelioration of DNA repair efficiency and decreased apoptosis. In line with these findings, genes involved in DNA repair and signaling pathways were found downregulated, whereas genes implicated in apoptosis were found upregulated in quiescent SLE patients.

Methods Patients

Peripheral blood mononuclear cells (PBMCs) from 12 patients (10 women) who fulfilled American College of

Page 2 of 12

Rheumatology criteria for SLE and had high serum titers of anti-double-stranded DNA autoantibodies at the time of sampling were studied. Six patients (all women, aged between 24 and 45 years, with disease duration ranging between 2 and 11 years) were selected on the basis of the presence of clinically quiescent disease for at least 6 months prior to sampling (SLE disease activity index Selena modification score of 6 or less, in the absence of any lupus-related clinical manifestation) under daily therapy with hydroxychloroquine only (n = 3), or in combination with 5 mg or less of prednisolone (n = 3). Six other patients (four women, aged between 21 and 59 years, with disease duration ranging between 1 and 15 years) were selected on the basis of the presence of proliferative glomerulonephritis; their characteristics, disease status, and medications have been described in detail elsewhere [13]. Twelve apparently healthy individuals, who were antinuclear antibody negative and matched 1:1 for age and gender to the patients, were studied in parallel. Cell cultures

Freshly isolated PBMCs were stimulated into proliferation using phytohemagglutinin (PHA; 10 μg/ml) for 48 hours and then treated with 100 μg/ml melphalan for 5 min at 37 °C in complete RPMI-1640 medium supplemented with 10 % fetal bovine serum (FBS), 100 units/ ml penicillin, 100 μg/ml streptomycin and 2 mmol/l Lglutamine. Cells were subsequently incubated in drugfree medium for various times (0–48 h), harvested, and stored at −70 °C. In some experiments, PBMCs were exposed to 2.5 μg/ml of vorinostat (suberoylanilide hydroxamic acid; Millipore Sigma, St. Louis, MO, USA) for 24 h [14], or 10 μg/ml a-amanitin (Millipore Sigma) for 6 h [15], treated with melphalan as above, harvested and stored at −70 °C. Nucleotide excision repair measurement

The N-ras gene-specific nucleotide excision repair was evaluated by the monofunctional binding of the drug melphalan to a single site in the DNA molecule (monoadducts) at various time points, as described elsewhere [16]. DNA double-strand breaks repair measurement

For the evaluation of DNA double-strand breaks the immunofluorescence antigen staining and confocal laser scanning microscope analysis was used as described previously [13]. Briefly, aliquots of 2 × 104 PBMCs were stimulated into proliferation using PHA, treated as described above, adhered to coverslip, fixed and stored at −70 °C until the analysis of γH2AX and Rad51. Cells were incubated with antibodies against γH2AX (serine139, Cell Signaling, Danvers, MA, USA) or Rad51 (Santa

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

Cruz Biotechnology, Dallas, TX, USA), washed, incubated with fluorescent secondary antibodies (Alexa Fluor 488 goat anti-mouse IgG; Abcam, Cambridge, UK) and images were visualized with a Leica TCS SP-1 confocal laser scanning microscope (Leica, Wetzlar, Germany). Foci were manually counted in 200 cells/treatment condition and results were expressed as the mean γH2AX or Rar51 foci per cell [mean ± standard deviation (SD)] from three independent experiments. Micrococcal nuclease digestion-based analysis of chromatin condensation

Nuclei were isolated from freshly isolated, untreated PBMCs (5 × 104) and digested with 1 unit of micrococcal nuclease (Millipore Sigma) for 0, 1, 2, 5 and 10 min at 37 °C. DNA was purified, separated in 1.5 % agarose gel, transferred onto nitrocellulose, and hybridized with the appropriate labeled probe [17]. Western blot analysis

Cells were harvested after treatment with various doses of vorinostat for 24 hours at 37 °C, rinsed in ice-cold phosphate-buffered saline (PBS), and lysed in buffer containing 50 mM HEPES (pH 7.9), 0.4 M NaCl, 1 mM EDTA, 2 μg/ml leupeptin, 2 μg/ml aprotinin, 5 μg/ml benzamidine, 0.5 mM phenylmethylsulfonyl fluoride (PMSF), and 1 % NP-40, followed by SDS-PAGE on 12 % polyacrylamide gels. Subsequently, the proteins were transferred to nitrocellulose membranes, which were incubated for 40 min at room temperature with nonfat dry milk (5 %) in 1 × Tris-buffered saline (TBS) followed by incubation with specific primary antibodies to nonacetylated histone H4, acetylated histone H4, and beta actin (Cell Signaling) overnight at 4 °C. Then, the membranes were washed with 0.3 % bovine serum albumin in 1 × TBS and incubated with horseradish peroxidase-conjugated secondary antibodies for 2 h at room temperature. After washing with 1 × TBS, the bound antibody complexes were visualized using the Western Blotting Detection Kit ECL Plus (Amersham, Little Chalfont, UK) and XOMAT-AR film (Kodak, Rochester, NY, USA). Apoptosis assay

Cells were treated with various doses of melphalan (0–200 μg/ml) for 5 min, followed by 24 h postincubation time. Then, the Cell Death Detection ELISA-PLUS kit (Roche Diagnostics Corp., Basel, Switzerland) was used to determine apoptosis according to the protocol provided by the manufacturer. RT2 Profiler™ PCR Array

Total RNA was extracted from freshly isolated untreated PBMCs using the Qiagen RNeasy Mini Kit (Qiagen,

Page 3 of 12

Venlo, the Netherlands) according to the manufacturer’s instructions and stored at −80 °C until use. For PCR array analysis, the RT2 Profiler™ PCR Array (Qiagen) of 84 genes related to the DNA damage signaling pathway was utilized. Total RNA (200 ng) was reverse transcribed using the RT2 First Strand kit following the manufacturer’s instructions. Resulting cDNA was diluted in nuclease-free water and added to RT2 SYBR Green Fluor qPCR Mastermix (Qiagen), which was subsequently aliquoted to each well of the PCR array for quantitative PCR. Thermal cycling and fluorescence detection were performed using a Biorad iQ™5 (BioRad Laboratories, Hemel Hempstead, UK). Cycling conditions were: 10 min denaturation at 95 °C followed by 40 cycles of 15 sec at 95 °C and 1 min at 60 °C. Six healthy control samples and six quiescent SLE patients samples were run on individual plates and data were analyzed using the RT2 Profiler PCR Array Data Analysis Webportal (http://pcrdataanaly sis.sabiosciences.com/pcr/arrayanalysis.php). Two independent experiments were performed for each sample. Systems biology statistical analysis

The transcripts presenting statistically significant differential expression (p < 0.05) were analyzed through the use of Qiagen’s Ingenuity® Pathway Analysis (IPA®, Qiagen, Redwood City, CA, USA, www.qiagen.com/in genuity) considering all direct and indirect relationships in the Ingenuity Knowledge base obtained only from experimentally verified information. Canonical pathways underwent enrichment analysis and the corresponding p values were calculated (p values < 0.05 were considered significant). Statistical analysis

The efficiency of DNA repair and the induction of apoptosis were compared between groups of individuals using the Wilcoxon rank sum test. Correlations between melphalan-induced DNA damage and induction of apoptosis were assessed by the linear regression analysis. A p value less than 0.05 was considered statistically significant. P values indicating statistical significance of the gene expression levels where corrected for multiple comparisons (FDR correction; Additional file 1: Table S1) [18].

Results Nucleotide excision repair and DNA double-strand breaks repair are defective in SLE, regardless of disease activity level

In the present study, two major DNA repair mechanisms were evaluated in SLE patients, i.e., nucleotide excision repair and DNA double-strand breaks repair. Based on our previous experience, we used the genotoxic drug melphalan as a tool to induce (a) DNA lesions such as

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

the monofunctional binding of this drug to a single site in the DNA molecule (monoadducts), which are almost exclusively repaired by nucleotide excision repair [16], and (b) DNA double-strand breaks, which are indirectly formed as a consequence of melphalan-induced oxidative stress [19] and as intermediates in the repair of melphalan-induced DNA damage [20, 21]. First, the efficiency of the nucleotide excision repair was measured in six patients with quiescent disease and their matched controls. Since we have previously shown that the efficiency of nucleotide excision repair at the Nras locus strongly correlates with melphalan-induced apoptosis in PBMCs from healthy controls [22], the repair kinetics of N-ras-specific monoadducts were followed for up to 48 h using Southern blot analysis (Fig. 1a). In all subjects, similar formation of monoadducts was found at the end of the 5-min melphalan treatment. Thereafter, their levels were decreased, with the removal capacity being higher in healthy controls than in quiescent SLE patients and lowest in patients with active proliferative nephritis (Fig. 1b). In accordance to these data, monoadducts burden (expressed as the area under the curve for DNA adducts during the whole experiment), a parameter strongly correlating with the cytotoxicity of genotoxic agents [23], was significantly higher in quiescent SLE patients compared to healthy controls; maximal values were observed in patients with active proliferative nephritis (Fig. 1c). Then, to study the formation and repair of DNA double-strand breaks, γH2AX foci as a marker of DNA damage and the formation of Rad51 foci as a marker of homologous recombination were determined. In all subjects analyzed, γH2AX foci reached maximal levels within 8 h; thereafter, their levels were decreased, with the removal capacity being higher in healthy controls than in SLE patients (Fig. 2a, b). Increased γH2AX levels were found in lupus nephritis compared to quiescent SLE patients; statistical significance was observed at 2 h and 48 h following melphalan treatment (Fig. 2b). In accordance to these results, higher γH2AX foci burden, expressed as AUC, was observed in quiescent SLE patients compared to healthy controls, whereas patients with active proliferative nephritis showed higher γH2AX burden than quiescent SLE patients (Fig. 2c). The Rad51 response followed the same time course as the γH2Ax response, peaking at 8 h and declining thereafter, with healthy controls showing lower Rad51 foci levels compared to quiescent SLE patients (Fig. 2d-f ).

Apoptosis rates of quiescent SLE-derived mononuclear cells correlate inversely with DNA repair efficiencies

The melphalan-induced apoptosis rates in PBMCs from quiescent SLE patients and their matched healthy

Page 4 of 12

Fig. 1 Impaired nucleotide excision repair in SLE patients. a Representative autoradiograms for the Southern blot analysis of melphalan-induced N-ras-specific DNA adducts. 0/0, no treatment. The kinetics of monoadducts (b) and total amounts of monoadducts expressed as AUC (c) in quiescent systemic lupus erythematosus (QSLE) and lupus nephritis (LN) patients as well as in 12 healthy controls (HC) are shown. The asterisks indicate significant differences (p < 0.05) between QSLE and LN patients. The experiments shown were based on a minimum of three independent repeats and the data reported are the mean ± SD of all subjects analyzed

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

Page 5 of 12

Fig. 2 Impaired double-strand breaks repair in quiescent SLE patients. a Typical images showing the γH2AX staining at different time points after treatment of PBMCs from one representative healthy control (HC) and one quiescent systemic lupus erythematosus (QSLE) patient after treatment with melphalan. Cell nuclei labeled with DAPI, γH2AX staining, as well as merged DAPI and γH2AX staining are shown. 0/0 no treatment. Formation of γH2AX foci (b) and total amounts of γH2AX foci expressed as AUC (c) after treatment of PBMCs from QSLE and lupus nephritis (LN) patients as well as from 12 HC with melphalan. The asterisks indicate significant differences (p < 0.05) between QSLE and LN patients. NSD no significant difference. d Typical images showing the Rad51 staining at different time points after treatment of PBMCs from one representative HC and one QSLE patient with melphalan. Cell nuclei labeled with DAPI, Rad51 staining, as well as merged DAPI and Rad51 staining are shown. 0/0 no treatment. Formation of Rad51 foci (e) and total amounts of Rad51 foci expressed as AUC (f) after treatment of PBMCs from QSLE patients and their matched HC with melphalan. The experiments shown were based on a minimum of three independent repeats and the data reported are the mean ± SD of all subjects analyzed

controls were also measured. We found that the lowest concentrations of melphalan required for the induction of apoptosis were significantly higher in healthy controls than in quiescent SLE patients (Fig. 3a), indicating that PBMCs from quiescent SLE patients are characterized

by increased apoptotic rates. Interestingly, an inverse correlation was observed between the melphalan concentration required for the induction of apoptosis and the levels of both types of DNA damage examined in the same quiescent SLE patients (linear regression analysis;

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

Page 6 of 12

Fig. 3 Increased apoptosis rates of quiescent SLE-derived mononuclear cells correlate with defective DNA repair efficiencies. a The induction of apoptosis after treatment of PBMCs from quiescent systemic lupus erythematosus (SLE) patients and their matched healthy controls (HC) with melphalan. Correlation between the melphalan doses required for the induction of apoptosis and the drug-induced monoadducts (b) or doublestrand breaks levels (c) in the same QSLE patients or healthy controls. d Correlation between individual efficiencies of nucleotide excision repair versus double-strand breaks repair (expressed as AUC) in all SLE patients (six with quiescent disease and six with proliferative nephritis) and 12 healthy controls. The experiments shown were based on a minimum of three independent repeats and the data reported are the mean ± SD of all the subjects analyzed

monoadducts: r2 = 0.831 for quiescent SLE patients and 0.920 for their matched controls, Fig. 3b; double-strand breaks: r2 = 0.878 for quiescent SLE patients and 0.907 for healthy controls, Fig. 3c). Notably, a strong correlation was found between the individual efficiencies of the two DNA repair mechanisms under study in all subjects analyzed (linear regression analysis: r2 = 0.767 for SLE patients and 0.567 for healthy controls, Fig. 3d). Defective chromatin organization and nucleotide excision repair in quiescent SLE are partially corrected by vorinostat

We next analyzed chromatin condensation in the N-ras gene locus, using micrococcal nuclease digestion of untreated PBMCs from quiescent SLE patients and their matched controls. As shown in Fig. 4a and b, patients are characterized by more condensed chromatin at the N-ras locus than their matched controls. That is, in healthy controls the N-ras gene locus gave rise mostly to mono- and di-nucleosome structures, with a significant portion in mono-nucleosomes, while the same locus in quiescent SLE patients gave rise to di-, tri-nucleosomes and higher structures. Chromatin organization and the efficiency of nucleotide excision repair were also evaluated in the N-ras

gene locus following treatment of PBMCs from quiescent SLE with vorinostat, a histone deacetylase inhibitor which has been approved by the US Food and Drug Administration for the treatment of refractory cutaneous T-cell lymphoma [24]. First, we determined the degree of H4 histone acetylation in PBMCs from quiescent SLE patients after treatment with vorinostat using Western blot analysis; nonacetylated histone H4 was included as a control for acetylated histone H4. We found that before incubation with vorinostat, the levels of acetylated histone H4 were low and did not increase remarkably with up to 1 μM vorinostat for 24 hours. However, incubation with 2.5 μM vorinostat for 24 h resulted in the maximal accumulation of acetylated histone H4 (Fig. 4d) associated with chromatin decondensation (Fig. 4c). In line with previous data showing that the rate of repair of UV-induced DNA damage increased following histone hyperacetylation by sodium butyrate [25], we found that vorinostat treatment of PBMCs from quiescent SLE patients resulted in increased efficiency of nucleotide excision repair (Fig. 4e) and decreased DNA damage burden of these cells (Fig. 4f ). Conversely, following treatment of PBMCs from quiescent SLE patients with a-amanitin, an inhibitor of RNA polymerase II, which causes conversion of chromatin into a more

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

condensed form [16], chromatin compaction of the N-ras gene locus (Additional file 1: Figure S1a, b) was accompanied by reduction of the efficiency of nucleotide excision repair and increased DNA damage burden (Additional file 1: Figure S1c, d). Vorinostat ameliorates DNA double-strand breaks repair efficiency and decreases apoptosis rates in quiescent SLE

The effects of the chromatin organization modifiers on the efficiency of double-strand breaks repair and the apoptosis rates were also evaluated. In line with previous data showing that the compaction of chromatin influences the efficiency of the DNA damage response and the sensitivity of cells to DNA double-strand breaks [26], we found that vorinostat treatment of PBMCs from quiescent SLE patients increased the repair efficiency of double-strand breaks (Fig. 4g), reduced double-strand breaks burden (Fig. 4h), and suppressed apoptosis rates of these cells (Fig. 4i). Conversely, in all subjects, treatment of PBMCs with a-amanitin resulted in higher double-strand breaks levels and increased apoptosis rates (Additional file 1: Figure S1e-g). Under the conditions used in this study, vorinostat or a-amanitin alone did not induce apoptosis in PBMCs from all subjects examined (data not shown). Altered expression of DNA repair- and apoptosis-related genes in quiescent SLE

The expression of genes involved in DNA damage repair and apoptosis pathways was analyzed in untreated PBMCs by RT2 Profiler™ PCR Array (Fig. 5a). Of 84 genes examined, a total of 37 genes representing several non-mutually exclusive categories demonstrated at least twofold difference in gene expression between healthy controls and quiescent SLE patients (Additional file 1: Tables S2 and S3). Particularly, the downregulated genes were categorized into nucleotide excision repair (DDB1, ERCC2, XPA, XPC), double-strand breaks repair (BLM, CHEK1, HUS1, MRE11A, NBN, RAD50, RAD51, RPA1, TP53BP1, XRCC2, XRCC6), base excision repair (LIG1, PARP1, XRCC1), mismatch repair (MLH1, MLH3) and signaling pathways (ATM, RAD1) (Fig. 5b). The expression of the DNA repair XRCC3 gene was also found decreased in SLE patients. On the other hand, several overexpressed genes are implicated in the apoptosis pathway, including PPP1R15A, CDKN1A, BARD1, RAD21, RAD9A, PRKDC, CIB1, BRCA1, ABL1, CHEK2 and BBC3 (Fig. 5c). Also, GADD45A, involved in DNA repair, and H2AFX, a DNA damage response gene, were overexpressed in lupus PBMCs. Finally, we performed a systems biology analysis of the significantly differentially expressed transcripts on canonical pathways through IPA®, which revealed

Page 7 of 12

significant involvement (enrichment p value < 0.05) of several DNA damage response and repair pathways, including nucleotide excision repair (p = 1.00E-007; Additional file 1: Figure S2), homologous recombination (p = 5.01E-020; Additional file 1: Figure S3), nonhomologous end joining (p = 2.51E-017; Additional file 1: Figure S4), base excision repair (p = 2.63E-010; Additional file 1: Figure S5), ATM signaling (p = 1.58E-043; Additional file 1: Figure S6), mismatch repair (p = 3.16E-014; Additional file 1: Figure S7), as well as apoptosis pathway signaling (p = 3.16E-003). Given that the enrichment analysis was performed on the differentially expressed genes only, the fact that most of the preselected genes were differentially expressed enhances the initial working hypothesis.

Discussion Herein, two major DNA repair mechanisms were evaluated ex vivo in patients with quiescent SLE and active lupus nephritis. The first mechanism, namely nucleotide excision repair was found to be less efficient in SLE patients compared to healthy controls, whereas patients with quiescent disease showed higher repair efficiency than those with active nephritis. Since the specific chromatin environment of a DNA lesion is considered an important factor that may affect recognition of lesions and repair rates [16, 27], a possible explanation for the lower efficiency of nucleotide excision repair could be related to a defective chromatin organization in our patients. Indeed, we found that quiescent SLE patients are characterized by more condensed local chromatin than healthy controls, which possibly impedes the access of DNA repair proteins to sites of DNA damage, thus reducing DNA repair capacity [28]. Whether a defective chromatin organization in SLE is associated with aberrations in histone acetylation processes remains unclear. In general, histone acetyltransferases and deacetylases regulate chromatin accessibility by adding or removing, respectively, acetyl groups to lysine residues in the N-terminal histone domains [29]. Acetylation neutralizes the positive charge of histone lysines, weakening electrostatic DNA-histone interactions and increasing DNA accessibility, while deacetylation strengthens DNA-histone interactions, thus decreasing DNA accessibility. Regarding the patterns of histone modifications in SLE patients, controversial results have been reported. For example, Zhou et al. [30] reported that histone H3 acetylation of the TNFSF7 (CD70) promoter in SLE CD4 + T cells is significantly increased, and may contribute to the development of lupus by increasing the expression of the CD70 gene. In addition, in monocytes isolated from SLE patients, Zhang et al. [31] identified a group of 179 genes with significantly enriched acetylation of H4 promoter compared to controls. Other data showed significantly increased total histone H4

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

Page 8 of 12

Fig. 4 Effects of vorinostat treatment of quiescent SLE-derived cells. Autoradiograms showing micrococcal nuclease sensitivity of the N-ras locus in untreated PBMCs from six healthy controls (HC) (a) and six quiescent systemic lupus erythematosus (QSLE) patients (b), as well as in PBMCs from the same QSLE patients after treatment with 2.5 μM vorinostat for 24 h (c). Symbols M, D and T represent the positions of nucleosome monomer, dimer and trimer respectively. d Western blot analysis of PBMCs from a representative quiescent SLE patient treated with various doses of vorinostat for 24 h. Nonacetylated histone H4 was used as a control for acetylated histone H4, and beta actin as a loading control. The kinetics of monoadducts (e) and γH2AX foci (g) as well as total amounts of monoadducts (f) and γH2AX foci expressed as AUC (h) after treatment of PBMCs from QSLE patients and their matched healthy controls with melphalan in the presence or not of vorinostat. The asterisks indicate significant differences (p < 0.05) between QSLE and LN patients. i The induction of apoptosis after treatment of PBMCs from the same QSLE patients and their matched HC with melphalan in the presence or not of vorinostat. The experiments shown were based on a minimum of three independent repeats and the data reported are the mean ± SD of all the subjects analyzed.

acetylation in SLE monocytes [32]. In contrast to the above, and in line with our findings, global hypoacetylation of histone H3 and H4 was observed in active lupus CD4+ T cells compared with controls [33]. Notably, the degree of histone H3 acetylation correlated inversely with SLE disease activity [33]. The second major mechanism studied herein, namely DNA double-strand breaks repair, was also found less efficient in SLE patients compared to healthy controls,

with the DNA double-strand breaks burden being higher in patients with active nephritis compared to those with quiescent SLE. These results are in line with recent data showing that both T cells and monocytes in SLE patients have increased sensitivity to acquire double-strand breaks following oxidative stress, which associates with disease activity [34]. Although we did not address the mechanistic basis for the decreased double-strand breaks repair efficiency of our patients, previous studies have

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

Page 9 of 12

Fig. 5 Expression of 84 genes involved in DNA damage response pathways. a Hierarchical clustergram of the 84 genes under study in untreated PBMCs from six quiescent systemic lupus erythematosus (QSLE) patients versus their matched healthy controls (HC). b, c Genes demonstrating at least twofold difference in the transcription activity between QSLE patients and their matched HC. Gene acronyms are explained in Additional file 1: Tables S2 and S3

shown that many repair proteins implicated in this repair mechanism are targets for autoantibodies. For example, autoantibodies against DNA repair proteins involved in nonhomologous end joining, including the two subunits of Ku protein, the poly(ADP-ribose) polymerase 1 (PARP-1) protein, the catalytic subunit of the DNAdependent protein kinase (DNA-PK) and the DNA ligase IV were all identified in patients with systemic autoimmune diseases [35–38]. Moreover, a cell-penetrating lupus autoantibody, 3E10, was found to preferentially bind DNA single-strand tails and inhibit key steps in DNA single- and double-strand breaks repair process [39]. Interestingly, we found that the individual capacities of both nucleotide excision repair and double-strand breaks repair mechanisms strongly correlated with the corresponding PBMCs apoptosis rates, in either SLE patients or healthy controls, supporting our hypothesis that accumulation of DNA damage contributes to increased apoptosis.

Following our assumption that the more condensed chromatin in our quiescent lupus patients is linked to deficient DNA repair, we considered that chemical modifiers of chromatin organization would affect repair efficiency. Indeed, we found that treatment of PBMCs from quiescent SLE patients with the histone deacetylase inhibitor vorinostat resulted in hyperacetylation of histone H4, chromatin decondensation, amelioration of both nucleotide excision repair and DNA double-strand breaks repair efficiencies, and decreased apoptosis. Previous studies have shown that the in vivo administration of several histone deacetylase inhibitors including vorinostat, corrected the site-specific hypoacetylation states on H3 and H4 lupus mouse models with marked improvement of disease phenotype [40–42]. In line with the data presented above, gene expression results showed that many genes involved in DNA damage repair and signaling pathways were downregulated in quiescent SLE patients, whereas, several genes

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

implicated in the apoptosis pathway were upregulated. To the best of our knowledge, this is the first study in SLE patients showing underexpression of the XPA, XPC, HUS1, MRE11A, NBN, RAD50, RAD51, RPA1, TP53BP1 and ATM genes involved in DNA damage repair and signaling pathways, as well as overexpression of PPP1R15A, BARD1, BBC3, BRCA1 genes involved in the apoptosis pathway. Moreover, in accordance to previous studies, we found downregulation of the excision repair cross-complementing ERCC2 and ERCC5 genes [43, 44], the X-ray repair cross-complementing XRCC6 gene [44], the DDB1 gene coding for the damage-specific DNA binding protein 1 [45], the Bloom syndrome RecQ helicase-like gene (BLM) [46], the XRCC1 gene [11] and the poly (ADP-ribose) polymerase 1 gene (PARP-1) [47]. Also, we found that SLE cells showed higher GADD45A mRNA levels as compared with healthy controls [48]. Overexpression of several other apoptosis-related genes has also been reported in PBMCs from SLE patients [49]. Regarding the pathogenetic mechanisms of autoimmunity, an important link between DNA damage repair and the activation of the type I interferon (IFN) characterizing SLE emerges [50, 51]. Indeed, Gunther et al. [52] reported recently that rare variants of the genome surveillance enzyme ribonuclease H2 gene are associated with systemic autoimmunity and that these variants resulted in impaired ribonucleotide removal. They also showed that such embedded ribonucleotides caused increased UV-induced cyclobutane pyrimidine dimer formation and enhanced type I IFN signaling, thus linking mutations in a DNA repair enzyme with systemic autoimmunity. Furthermore, Härtlova et al. [53] showed that accumulation of spontaneous DNA lesions stimulates type I IFN system via the activation of the STING pathway by self-DNA released into the cytoplasm, implying that the DNA repair network emerges as an important new player in the pathogenesis of autoimmunity. Moreover, a polymorphism in the DNA repair gene RAD51B has been associated with increased risk of SLE [54]. A limitation of the present work is the small sample size of patients with quiescent SLE; increasing the number of patients would increase confidence in the data generated. However, the main conclusion of this study, i.e., that both nucleotide excision repair and DNA double-strand breaks repair were defective in SLE patients (a) was confirmed in both quiescent SLE and in lupus nephritis patients (12 SLE patients versus 12 healthy controls) and (b) is in line with previous data showing that SLE T cells and monocytes have increased sensitivity to acquiring DNA double-strand breaks in response to oxidative stress [34]. Moreover,

Page 10 of 12

since total PBMCs were used in the present study, we cannot exclude the possibility that differences in cell subsets constituents between patients and controls contribute to some of the differences observed. Notably, a recent report provides evidence for defective repair of oxidative stress-induced double-strand breaks in all peripheral blood mononuclear cell subsets (CD4+ T cells, CD8+ T cells and monocytes) derived from SLE patients compared with those from healthy controls [34]. Also, previous studies have also shown that both lymphocytes and neutrophils from SLE patients display increased DNA damage and, additionally, may demonstrate defective repair of oxidative DNA damage [9, 10].

Conclusions Our findings support the hypothesis that increased apoptosis in SLE could be due, at least in part, to defective DNA repair and chromatin organization (Fig. 6). Based on our findings we propose that SLE patients, for reasons currently unknown, are characterized by condensed chromatin in certain loci, resulting in lower repair efficiency of important DNA repair pathways (nucleotide excision repair and DNA double-strand breaks repair) and, thus, increased DNA damage burden, which in turn triggers the induction of the apoptotic pathway. Although not tested in vivo, i.e., in lupus mouse models, treatment of human SLE-

Fig. 6 Epigenetically regulated functional abnormalities of DNA repair machinery may promote lymphocyte apoptosis in SLE: a proposed model (for details see “Conclusions”)

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

derived cells with the histone deacetylase inhibitor vorinostat results in chromatin decondensation, amelioration of DNA repair efficiency, and decreased apoptosis. Therefore, further work on the mechanisms of the epigenetic imbalance in DNA damage repair may reveal potential therapeutic targets in SLE.

Additional file Additional file 1: Table S1. Gene expression levels corrected for multiple comparisons (FDR correction). Table S2. Genes that were downregulated in quiescent SLE patients. Table S3. Genes that were upregulated in quiescent SLE patients. Figure S1. Effect of a-amanitin treatment of quiescent SLE-derived cells. Figure S2. Canonical pathways related to DNA damage response and repair from IPA® presenting statistically significant (p < 0.05) enrichment: nucleotide excision repair (p = 1.00E-007). Figure S3. Canonical pathways related to DNA damage response and repair from IPA® presenting statistically significant (p < 0.05) enrichment: DNA double-strand breaks repair by homologous recombination (p = 5.01E-020). Figure S4. Canonical pathways related to DNA damage response and repair from IPA® presenting statistically significant (p < 0.05) enrichment: DNA double-strand breaks repair by nonhomologous end joining (p = 2.51E-017). Figure S5. Canonical pathways related to DNA damage response and repair from IPA® presenting statistically significant (p < 0.05) enrichment: base excision repair (p = 2.63E-010). Figure S6. Canonical pathways related to DNA damage response and repair from IPA® presenting statistically significant (p < 0.05) enrichment: ATM signaling (p = 1.58E-043). Figure S7. Canonical pathways related to DNA damage response and repair from IPA® presenting statistically significant (p < 0.05) enrichment: mismatch repair (p = 3.16E-014). (DOC 7236 kb) Abbreviations FBS, fetal bovine serum; IFN, interferon; PBMCs, peripheral blood mononuclear cells; PBS, phosphate-buffered saline; PHA, phytohemagglutinin; PMSF, phenylmethylsulfonyl fluoride; SLE, systemic lupus erythematosus; TBS, Tris-buffered saline Acknowledgements We would like to thank Prof. George C. Tsokos (Division of Rheumatology, Harvard Medical School) for critical review of the manuscript. Funding The study was funded by the Special Account for Research Grants of the National and Kapodistrian University of Athens, Athens, Greece (ELKE Grant number 974). Availability of supporting data The datasets supporting the conclusions of this article are available in the Helios Digital Repository: http://helios-eie.ekt.gr/EIE/handle/10442/15330. Authors’ contributions VLS and PPS directed and coordinated the project, performed data acquisition and data analysis, and drafted the manuscript. KV and VGG contributed to data analysis and revised the manuscript. All authors read and approved the final manuscript. Authors’ information VLS: Research Associate Professor, Institute of Biology, Medicinal Chemistry and Biotechnology (IBMCB), National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Ave., 11635 Athens, Greece. KV: Staff Research Scientist-Associate Professor Level, Biomedical Research Foundation of the Academy of Athens (BRFAA), 4 Soranou Ephessiou St., 115 27 Athens, Greece. VGG: Professor, Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Medical School National and Kapodistrian University of Athens and Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.

Page 11 of 12

PPS: Professor, First Department of Prop. Internal Medicine and Rheumatology Unit, Joint Academic Rheumatology Program, National Kapodistrian University of Athens Medical School, Athens, Greece. Competing interests The authors declare that they have no competing interests. Consent for publication Not applicable. Ethical approval and consent to participate The study was approved by the Laikon Hospital ethics committee and all participants provided informed consent according to the Declaration of Helsinki. Author details 1 Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece. 2Joint Rheumatology Program and First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece. 3 Biomedical Research Foundation, Academy of Athens, Athens, Greece. 4 Molecular Carcinogenesis Group, Department of Histology and Embryology, National and Kapodistrian University of Athens Medical School, Athens, Greece. Received: 16 May 2016 Accepted: 21 July 2016

References 1. Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365:2110–21. 2. Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell. 2010;140:619–30. 3. Bertsias GK, Salmon JE, Boumpas DT. Therapeutic opportunities in systemic lupus erythematosus: state of the art and prospects for the new decade. Ann Rheum Dis. 2010;69:1603–11. 4. Arbuckle MR, Mc Clain MT, Rubertone MV, Scofield RH, Dennis GJ, James JA, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003;349:1526–33. 5. Ohnishi T, Mori E, Takahashi A. DNA double-strand breaks: their production, recognition, and repair in eukaryotes. Mutat Res. 2009;669:8–12. 6. Sirbu BM, Cortez D. DNA damage response: three levels of DNA repair regulation. Cold Spring Harb Perspect Biol. 2013;5:a012724. 7. Friedberg EC. How nucleotide excision repair protects against cancer. Nat Rev Cancer. 2001;1:22–33. 8. Helleday T, Lo J, van Gent DC, Engelward BP. DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair (Amst). 2007;6:923–35. 9. Bashir S, Harris G, Denman MA, Blake DR, Winyard PG. Oxidative DNA damage and cellular sensitivity to oxidative stress in human autoimmune diseases. Ann Rheum Dis. 1993;52:659–66. 10. McConnell JR, Crockard AD, Cairns AP, Bell AL. Neutrophils from systemic lupus erythematosus patients demonstrate increased nuclear DNA damage. Clin Exp Rheumatol. 2002;20:653–60. 11. Bassi C, Xavier DJ, Palomino G, Nicolucci P, Soares C, Sakamoto-Hojo E, et al. Efficiency of the DNA repair and polymorphisms of the XRCC1, XRCC3 and XRCC4 DNA repair genes in systemic lupus erythematosus. Lupus. 2008;17: 988–95. 12. Davies RC, Pettijohn K, Fike F, Wang J, Nahas SA, Tunuguntla R, et al. Defective DNA double-strand break repair in pediatric systemic lupus erythematosus. Arthritis Rheum. 2012;64:568–78. 13. Souliotis VL, Sfikakis PP. Increased DNA double-strand breaks and enhanced apoptosis in patients with lupus nephritis. Lupus. 2015;24:804–15. 14. Munshi A, Tanaka T, Hobbs ML, Tucker SL, Richon VM, Meyn RE. Vorinostat, a histone deacetylase inhibitor, enhances the response of human tumor cells to ionizing radiation through prolongation of γ-H2AX foci. Mol Cancer Ther. 2006;5:1967–74. 15. Souliotis VL, Dimopoulos MA, Episkopou HG, Kyrtopoulos SA, Sfikakis PP. Preferential in vivo DNA repair of melphalan-induced damage in human genes is greatly affected by the local chromatin structure. DNA Repair (Amst). 2006;5:972–85.

Souliotis et al. Arthritis Research & Therapy (2016) 18:182

16. Episkopou H, Kyrtopoulos SA, Sfikakis PP, Fousteri M, Dimopoulos MA, Mullenders LH, et al. Association between transcriptional activity, local chromatin structure, and the efficiencies of both subpathways of nucleotide excision repair of melphalan adducts. Cancer Res. 2009;69:4424–33. 17. Episkopou H, Kyrtopoulos SA, Sfikakis PP, Dimopoulos MA, Souliotis VL. The repair of melphalan-induced DNA adducts in the transcribed strand of active genes is subject to a strong polarity effect. Mutat Res. 2011; 714:78–87. 18. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Statist Soc Ser B (Methodological). 1995;57:289–300. 19. Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 2013;12:931–47. 20. Simonelli V, Narciso L, Dogliotti E, Fortini P. Base excision repair intermediates are mutagenic in mammalian cells. Nucleic Acids Res. 2005; 33:4404–11. 21. Deans AJ, West SC. DNA interstrand crosslink repair and cancer. Nat Rev Cancer. 2011;11:467–80. 22. Gkotzamanidou M, Terpos E, Bamia C, Kyrtopoulos SA, Sfikakis PP, Dimopoulos MA, et al. Progressive changes in chromatin structure and DNA damage response signals in bone marrow and peripheral blood during myelomagenesis. Leukemia. 2014;28:1113–21. 23. Hansson J, Lewensohn R, Ringborg U, Nilsson B. Formation and removal of DNA cross-links induced by melphalan and nitrogen mustard in relation to drug-induced cytotoxicity in human melanoma cells. Cancer Res. 1987;47: 2631–7. 24. Hopfinger G, Nösslinger T, Lang A, Linkesch W, Melchardt T, Weiss L, et al. Lenalidomide in combination with vorinostat and dexamethasone for the treatment of relapsed/refractory peripheral T cell lymphoma (PTCL): report of a phase I/II trial. Ann Hematol. 2014;93:459–62. 25. Smerdon MJ, Lan SY, Calzal RE, Reeve R. Sodium butyrate stimulates DNA repair in UV-irradiated normal and xeroderma pigmentosum human fibroblasts. J Biol Chem. 1982;257:13441–7. 26. Murga M, Jaco I, Fan Y, Soria R, Martinez-Pastor B, Cuadrado M, et al. Global chromatin compaction limits the strength of the DNA damage response. J Cell Biol. 2007;178:1101–8. 27. Feng Z, Hu W, Chasin LA, Tang MS. Effects of genomic context and chromatin structure on transcription-coupled and global genomic repair in mammalian cells. Nucleic Acids Res. 2003;31:5897–906. 28. Gong F, Kwon Y, Smerdon MJ. Nucleotide excision repair in chromatin and the right of entry. DNA Repair (Amst). 2005;4(10):884–96. 29. Patel DR, Richardson BC. Epigenetic mechanisms in lupus. Curr Opin Rheumatol. 2010;22:478–82. 30. Zhou Y, Qiu X, Luo Y, Yuan J, Li Y, Zhong Q, et al. Histone modifications and methyl-CpG-binding domain protein levels at the TNFSF7 (CD70) promoter in SLE CD4 + T cells. Lupus. 2011;20:1365–71. 31. Zhang Z, Song L, Maurer K, Petri MA, Sullivan KE. Global H4 acetylation analysis by ChIP-chip in systemic lupus erythematosus monocytes. Genes Immun. 2010;11:124–33. 32. Leung YT, Shi L, Maurer K, Song L, Zhang Z, Petri M, et al. Interferon regulatory factor 1 and histone H4 acetylation in systemic lupus erythematosus. Epigenetics. 2015;10:191–9. 33. Hu N, Qiu X, Luo Y, Yuan J, Li Y, Lei W, et al. Abnormal histone modification patterns in lupus CD4+ T-cells. J Rheumatol. 2008;35:804–10. 34. Namas R, Renauer P, Ognenovski M, Tsou PS, Sawalha AH. Histone H2AX phosphorylation as a measure of DNA double-strand breaks and a marker of environmental stress and disease activity in lupus. Lupus Sci Med. 2016;3: e000148. 35. Takeda Y, Dynan WS. Autoantibodies against DNA double strand break repair proteins. Front Biosci. 2001;6:1412–22. 36. Reeves WH. Antibodies to the p70/p80 (Ku) antigens in systemic lupus erythematosus. Rheum Dis Clin North Am. 1992;18:391–414. 37. Muller S, Briand JP, Barakat S, Lagueux J, Poirier GG, De Murcia G, et al. Autoantibodies reacting with poly(ADP-ribose) and with a zinc-finger functional domain of poly(ADP-ribose) polymerase involved in the recognition of damaged DNA. Clin Immunol Immunopathol. 1994;73: 187–96. 38. Lee KJ, Dong X, Wang J, Takeda Y, Dynan WS. Identification of human autoantibodies to the DNA ligase IV/XRCC4 complex and mapping of an autoimmune epitope to a potential regulatory region. J Immunol. 2002; 169:3413–21.

Page 12 of 12

39. Hansen JE, Chan G, Liu Y, Hegan DC, Dalal S, Dray E, et al. Targeting cancer with a lupus autoantibody. Sci Transl Med. 2012;4:157ra142. 40. Mishra N, Reilly CM, Brown DR, Ruiz P, Gilkeson GS. Histone deacetylase inhibitors modulate renal disease in the MRL-lpr/lpr mouse. J Clin Invest. 2003;111:539–52. 41. Waibel M, Christiansen AJ, Hibbs ML, Shortt J, Jones SA, Simpson I, et al. Manipulation of B-cell responses with histone deacetylase inhibitors. Nat Commun. 2015;6:6838. 42. Regna NL, Chafin CB, Hammond SE, Puthiyaveetil AG, Caudell DL, Reilly CM. Class I and II histone deacetylase inhibition by ITF2357 reduces SLE pathogenesis in vivo. Clin Immunol. 2014;151:29–42. 43. Frangou EA, Bertsias GK, Boumpas DT. Gene expression and regulation in systemic lupus erythematosus. Eur J Clin Invest. 2013;43:1084–96. 44. Lee HM, Sugino H, Aoki C, Nishimoto N. Underexpression of mitochondrialDNA encoded ATP synthesis-related genes and DNA repair genes in systemic lupus erythematosus. Arthritis Res Ther. 2011;13:R63. 45. O’Hanlon TP, Rider LG, Gan L, Fannin R, Paules RS, Umbach DM, et al. Gene expression profiles from discordant monozygotic twins suggest that molecular pathways are shared among multiple systemic autoimmune diseases. Arthritis Res Ther. 2011;13:R69. 46. Kaneko H, Fukao T, Kasahara K, Yamada T, Kondo N. Augmented cell death with Bloom syndrome helicase deficiency. Mol Med Rep. 2011;4:607–9. 47. Rapoport MJ, Bloch O, Amit-Vasina M, Yona E, Molad Y. Constitutive abnormal expression of RasGRP-1 isoforms and low expression of PARP-1 in patients with systemic lupus erythematosus. Lupus. 2011;20:1501–9. 48. Li Y, Zhao M, Yin H, Gao F, Wu X, Luo Y, et al. Overexpression of the growth arrest and DNA damage-induced 45alpha gene contributes to autoimmunity by promoting DNA demethylation in lupus T cells. Arthritis Rheum. 2010;62:1438–47. 49. Eggleton P, Harries LW, Alberigo G, Wordsworth P, Viner N, Haigh R, et al. Changes in apoptotic gene expression in lymphocytes from rheumatoid arthritis and systemic lupus erythematosus patients compared with healthy lymphocytes. J Clin Immunol. 2010;30:649–58. 50. Baccala R, Hoebe K, Kono DH, Beutler B, Theofilopoulos AN. TLR-dependent and TLR-independent pathways of type I interferon induction in systemic autoimmunity. Nat Med. 2007;13:543–51. 51. Pateras IS, Havaki S, Nikitopoulou X, Vougas K, Townsend PA, Panayiotidis MI, et al. The DNA damage response and immune signaling alliance: is it good or bad? Nature decides when and where. Pharmacol Ther. 2015;154: 36–56. 52. Gunther C, Kind B, Reijns MA, Berndt N, Martinez-Bueno M, Wolf C, et al. Defective removal of ribonucleotides from DNA promotes systemic autoimmunity. J Clin Invest. 2015;125:413–24. 53. Härtlova A, Erttmann SF, Raffi FA, Schmalz AM, Resch U, Anugula S, et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity. 2015; 42:332–43. 54. Bentham J, Morris DL, Cunninghame Graham DS, Pinder CL, Tombleson P, Behrens TW, et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat Genet. 2015;47:1457–64.

Submit your next manuscript to BioMed Central and we will help you at every step: • We accept pre-submission inquiries • Our selector tool helps you to find the most relevant journal • We provide round the clock customer support • Convenient online submission • Thorough peer review • Inclusion in PubMed and all major indexing services • Maximum visibility for your research Submit your manuscript at www.biomedcentral.com/submit