• Nbs1 is a component of the MRE11 complex, which is a sensor of DNA double-strand breaks and plays a crucial role in the DNA damage response.

  • In mice with a hypomorphic allele of Nbs1, macrophages exhibit increased senescence and abnormal proliferation and inflammatory responses.

Nijmegen breakage syndrome 1 (NBS1) is a component of the MRE11 complex, which is a sensor of DNA double-strand breaks and plays a crucial role in the DNA damage response. Because activated macrophages produce large amounts of reactive oxygen species (ROS) that can cause DNA lesions, we examined the role of NBS1 in macrophage functional activity. Proliferative and proinflammatory (interferon gamma [IFN-γ] and lipopolysaccharide [LPS]) stimuli led to increased NBS1 levels in macrophages. In mice expressing a hypomorphic allele of Nbs1, Nbs1∆B/∆B, macrophage activation–induced ROS caused increased levels of DNA damage that were associated with defects in proliferation, delayed differentiation, and increased senescence. Furthermore, upon stimulation, Nbs1∆B/∆B macrophages exhibited increased expression of proinflammatory cytokines. In the in vivo 2,4-dinitrofluorobenezene model of inflammation, Nbs1∆B/∆B animals showed increased weight and ear thickness. By using the sterile inflammation by zymosan injection, we found that macrophage proliferation was drastically decreased in the peritoneal cavity of Nbs1∆B/∆B mice. Our findings show that NBS1 is crucial for macrophage function during normal aging. These results have implications for understanding the immune defects observed in patients with NBS and related disorders.

Endogenous sources of DNA damage, such as DNA replication or reactive oxygen species (ROS), can cause a diverse spectrum of DNA lesions.1  Among these, DNA double-strand breaks (DSBs) are considered one of the most cytotoxic and can promote oncogenic translocations if not properly metabolized. DSBs are repaired by 2 major pathways: nonhomologous end-joining and homologous recombination.2,3  The MRE11 complex, composed of meiotic recombination 11 homolog (MRE11), Nijmegen breakage syndrome 1 (NBS1), and RAD50, is a DSB sensor that regulates the DNA damage response (DDR) and repair of DSBs.4  Break detection by the MRE11 complex activates the ataxia telangiectasia mutated (ATM) kinase that promotes a robust DDR that includes the activation of checkpoint kinase 2 (CHK2) and the tumor suppressor p53.5 

NBS1 has no identifiable enzymatic activities and appears to function primarily as an adaptor protein required for MRE11 complex nuclear localization. NBS1 has a forkhead-associated domain and two BRCA1 C-terminal (BRCT) domains involved in phospho-dependent protein interactions in its N terminus and an MRE11 and PI3K-related protein kinase (PIKK) binding domain in its C terminus.6,7  The latter domain interacts with ATM and is required for an efficient apoptotic response in the immune system.7,8 

Mutations in any MRE11 complex members or ATM underlies rare genetic instability syndromes with overlapping pathologies that affect the central nervous system, germline, and immune system.4,7 NBS1 mutations cause Nijmegen breakage syndrome (NBS),7,9,10  characterized by microcephaly, growth and mental retardation, immunodeficiency, and predisposition to cancer.11  This syndrome highlights the importance of the DDR in the development and function of the central nervous and immune systems.

Immunological defects in NBS patients are poorly understood. T and B cells have been the major focus, because variable (diverse) joining segments and class switch recombination result in the production and repair of DSBs to facilitate immunoglobulin development.12,13 

Macrophages play a critical role during proinflammatory (destructive) and anti-inflammatory (repairing) phases of inflammation.14-16  Recently, it has been found that when an inflammation is produced, Ly6Chigh monocytes are released by the bone marrow (BM) to fight pathogenic intruders at inflammatory loci throughout the body.17  Macrophages may experience DSBs during the proliferation and also during activation, because they are the major producer of NO and ROS that can induce these and other lesions.18  This suggests that the proper control of DNA damage repair and proliferation is likely to be important in macrophages19  and could be affected by mutations in the MRE11 complex.

To address this possibility, we examined the role of NBS1 in macrophages using mice that express a hypomorphic allele of Nbs1, Nbs1∆B/∆B.20 Nbs1∆B/∆B mice express a truncated allele that lacks the N-terminal forkhead-associated and BRCT domains that are crucial for cell cycle checkpoints, DNA damage sensitivity, and efficient ATM signaling.8,20  Our results demonstrate that reduced NBS1 function promotes macrophage senescence, reflected by slow macrophage differentiation, reduced proliferation, and an impaired inflammatory response and suggest that its role in macrophages may contribute to the immunodeficiency observed in patients with NBS.

Mice

Nbs1∆B/∆B mice were a kind gift from Dr John H. Petrini.20  All mice were maintained in a specific-pathogen-free facility in the Parc Cientific de Barcelona. Animal use was approved by the Animal Research Committee of the Government of Catalonia, no. 2523.

Reagents

Murine recombinant interferon gamma (IFN-γ), macrophage colony-stimulating factor (M-CSF), granulocyte macrophage CSF (GM-CSF), and interleukin-4 (IL-4) were purchased from R&D Systems (Minneapolis, MN), and CpG-B was purchased from InvivoGen (San Diego, CA). All chemicals used were of the highest purity grade available from Sigma-Aldrich (St. Louis, MO), unless stated otherwise.

BM-derived macrophage culture

BM-derived macrophages (BMDMs) were generated as described.21  A homogeneous population of adherent macrophages was obtained after 7 days of culture (>99% CD11b and F4/80).

RNA extraction, qPCR, and PCR

Total RNA was extracted, purified, and DNase treated with PureLink RNA Mini Kit as recommended by the manufacturer (Ambion, Life Technologies). Complementary DNA synthesis and quantitative polymerase chain reaction (qPCR) were performed by using the SYBR Green Master Mix (Applied Biosystems) as described.22  Data were normalized using Hprt1 and/or l14 as housekeeping genes. Primer sequences are presented in supplemental Table 1, available on the Blood Web site. For PCR analysis of Nbs1 fragments, the total RNA extracted was used as a template for complementary DNA preparation, and the conditions of the PCR were 5 minutes at 95°C, 30 seconds at 95°C, 30 seconds at 58°C, and 1 minute at 72°C (for 30 cycles), plus a 10-minute extension at 72°C.

Western blot protein analysis

Western blotting was performed as described23  by using antibodies against NBS1 (Novus Biologicals), ATR (Cell Signaling), CHK2 (EMD Millipore), β-actin (Sigma-Aldrich), and LC3 (Sigma-Aldrich).

Telomere length

DNA was extracted by using the DNeEasy Blood and Tissue kit (Qiagen), and DNA was amplified by qPCR with telomere-specific primers and control primers (supplemental Table 1), as described.24 

Metaphase spreads

BMDMs were incubated with colcemid (0.1 μg/mL) for 30 minutes to 1 hour. BMDMs were swelled in 0.56% KCl for 15 minutes at 37°C and then fixed in ice-cold fixative (methanol and acetic acid, 3:1) and washed several times in fixative. Metaphase preparations were spread on precleaned glass slides, dried at room temperature, and stained with 4,6 diamidino-2-phenylindole in mounting medium.25  One hundred macrophages were scored per genotype.

Macrophage proliferation, cell cycle, and DNA content

Proliferation was measured as described.21  For cell cycle analysis, 106 cells were plated with 3 mL of 10% fetal calf serum Dulbecco’s modified Eagle medium and incubated for 16 hours before the addition of stimulants. Cells were collected after 24 hours, fixed with 95% ethanol, incubated with propidium iodide (PI) and RNAse-A, and later analyzed by flow cytometry.

Immunofluorescence

DNA breaks were quantified by counting the number of γH2AX foci per nuclei. For each condition, 10 different fields with at least 14 cells each were analyzed. The results are shown as the number of foci per nucleus in each field. The γH2AX antibody (EMD Millipore) was used with an Alexa-Fluor 488-labeled secondary antibody (Molecular Probes).

ROS

ROS production was measured by using dichlorofluorescin diacetate as described.24 

Sterile inflammation

Mice were injected intraperitoneally with 2 × 106 zymosan particles resuspended in phosphate-buffered saline (PBS) or with PBS alone.26  After 48 hours, mice were euthanized and peritoneal lavage was performed. The recovered cells were stained for extracellular CD11b and F4/80 prior to fixation and permeabilization. Cells were then stained for the intracellular proliferation marker Ki-67 with the PE Mouse Ki-67 Set (BD Biosciences). After washing, sample phenotypes were determined by flow cytometry.

Cutaneous inflammation

A solution of 0.5% 2,4-dinitrofluorobenezene (DNFB) in acetone was applied on the right ear of each mouse and acetone was applied on the left ear as a control.27  Animals were euthanized on day 7 after application. Ears were used for RNA and histology analysis after 24 hours of 4% paraformaldehyde fixation and paraffin embedding. Ear sections were stained with hematoxylin and eosin (H&E). Images were collected with a Nikon E800 microscope, and maximum ear thickness measurements were calculated with Fiji software.

Flow cytometry

BMDMs were collected and 5 × 105 cells were analyzed by using anti-CD115-PE (phycoerythrin) (eBioscience), anti-Ly6C-FITC (fluorescein isothiocyanate) (BD Pharmigen), anti-F4/80-APC (allophycocyanin) (eBioscience), anti-CD45-PECy7 (eBioscience), and anti-CD11b-BV711 (Biolegent) plus 4′,6 diamidino-2-phenylindole to exclude dead cells. To detect major histocompatibility complex (MHC) class II expression, an anti-MHC II-FITC (BD Pharmigen) was used. Samples were acquired on a Gallios Flow Cytometer (Beckman Coulter) and data were analyzed with FlowJo software (TreeStar).

Statistical analysis

Data were analyzed by using the nonparametric Mann-Whitney U test with GraphPad Prism 5 software.

NBS1 levels in macrophages are regulated by proinflammatory stimuli and M-CSF

Despite the high levels of DNA damage that occur in macrophages, the impact of attenuated MRE11 complex function has not been investigated.28,29  To address this, we first examined the expression of Nbs1 in macrophages to determine whether it was modulated by different stimulatory agents. We obtained murine BM and differentiated it in vitro to BMDMs. The resulting homogenous population of cells was composed of quiescent primary macrophages that could be induced to proliferate by growth factors or that could be classically and alternatively activated by cytokines.30  BMDMs were incubated with different proinflammatory (IFN-γ and LPS), anti-inflammatory (IL-4), and proliferative stimuli (M-CSF and GM-CSF), and levels of NBS1 were determined after treatment. No significant changes in Nbs1 messenger RNA (mRNA) levels (Figure 1A) were observed. However, after IFN-γ, LPS or M-CSF treatment, the levels of NBS1 protein were increased (Figure 1B). The changes in total NBS1 protein levels were specific, because the total levels of ATR, KU80, and CHK2 (other DSB-responsive proteins) were unaffected by macrophage stimulation (Figure 1C). To determine whether the increase was the result of the expression of alternative mRNA transcripts, we conducted a qualitative PCR analysis using primers that spanned the 2491-bp Nbs1 mRNA (NM_013752.3). In each case, we observed the same size and expression level, regardless of stimuli (supplemental Figure 1). Although the mechanism of protein stabilization during proinflammatory activation remained unclear, it suggested that NBS1 may have an important functional activity.

Figure 1

NBS1 is induced in macrophages after proinflammatory and M-CSF stimulation. Seven-day BMDMs were starved of growth factors for 16 to 18 hours and then incubated with different stimuli. IFN-γ (10 ng/mL) and LPS (10 ng/mL) were used as proinflammatory stimuli, IL-4 (10 ng/mL) as anti-inflammatory stimulus, and M-CSF (10 ng/mL) and GM-CSF (10 ng/mL) as growth factors. (A) Relative mRNA expression was determined at the indicated times after treatment. Each graph represents triplicate samples, and the results are shown as mean ± standard deviation (SD). (B) Protein levels of NBS1 were determined after 24 hours of stimulation. β-actin is included as a loading control. (C) Protein levels of other DNA damage response proteins were determined in macrophages upon stimulation with IFN-γ or M-CSF. β-actin is included as a loading control. (D) DNA damage was assessed by quantifying γH2AX foci. For each condition, 10 different fields with at least 14 cells each were quantified and the results are shown as number of foci per nucleus in each field. N-acetyl cysteine (NAC), a ROS scavenger, was used at 1 mM. All assays are representative of at least 3 independent experiments showing similar results. *P < .01 in relation to the controls when all the independent experiments had been compared.

Figure 1

NBS1 is induced in macrophages after proinflammatory and M-CSF stimulation. Seven-day BMDMs were starved of growth factors for 16 to 18 hours and then incubated with different stimuli. IFN-γ (10 ng/mL) and LPS (10 ng/mL) were used as proinflammatory stimuli, IL-4 (10 ng/mL) as anti-inflammatory stimulus, and M-CSF (10 ng/mL) and GM-CSF (10 ng/mL) as growth factors. (A) Relative mRNA expression was determined at the indicated times after treatment. Each graph represents triplicate samples, and the results are shown as mean ± standard deviation (SD). (B) Protein levels of NBS1 were determined after 24 hours of stimulation. β-actin is included as a loading control. (C) Protein levels of other DNA damage response proteins were determined in macrophages upon stimulation with IFN-γ or M-CSF. β-actin is included as a loading control. (D) DNA damage was assessed by quantifying γH2AX foci. For each condition, 10 different fields with at least 14 cells each were quantified and the results are shown as number of foci per nucleus in each field. N-acetyl cysteine (NAC), a ROS scavenger, was used at 1 mM. All assays are representative of at least 3 independent experiments showing similar results. *P < .01 in relation to the controls when all the independent experiments had been compared.

Close modal

To address this, we used mice that express a hypomorphic allele of Nbs1, Nbs1∆B/∆B.20 We observed increased numbers of γH2AX foci, a marker of DNA breaks, in macrophages from Nbs1∆B/∆B mice, and they were further increased by treatment with LPS, which induced increased levels of ROS31  (Figure 1D and supplemental Figure 2). To determine whether ROS was required for the increased DSBs observed in Nbs1∆B/∆B macrophages, we added N-acetyl cysteine, a ROS scavenger. In both the wild-type (WT) and Nbs1∆B/∆B macrophages, we observed a significant reduction in DSBs measured by γH2AX. This demonstrates that NBS1 is induced after macrophage activation and is critical for reducing DNA damage caused by increased ROS.

Nbs1 is required for M-CSF-dependent proliferation in macrophages

The MRE11 complex plays important roles in suppressing DNA damage during S phase.32  To examine the influence of the MRE11 complex on macrophage proliferation, we treated macrophages with M-CSF, which is crucial for both the differentiation and proliferation of macrophages.21  BM was extracted from 6- to 10-week-old WT and Nbs1∆B/∆B mice, and cells were counted prior to incubation with M-CSF media for differentiation. Although the total number of BM cells obtained from WT and Nbs1∆B/∆B mice did not differ, the amount of differentiated macrophages obtained from Nbs1∆B/∆B mice after 7 days was approximately half that obtained from the WT littermates (Figure 2A).

Figure 2

Reduced proliferation of macrophages from Nbs1∆B/∆B mice. (A) The number of total cells from the BM at day 0 of differentiation (BM after it was flushed from bones) and after 6 days of differentiation (in media containing M-CSF) from the indicated genotypes. (B) Macrophages were starved of growth factor for 18 hours, incubated for 1 hour with the indicated stimuli (LPS or IFN-γ), washed, and incubated for 24 hours with M-CSF (10 ng/mL) in a hyperoxic (20% O2) or hypoxic (1% O2) environment, and proliferation was quantified by incorporation of tritium. (C) Macrophages were starved of growth factor for 18 hours and incubated for 24 hours with M-CSF. Cell cycle distribution was determined by staining fixed cells with PI and assessment by flow cytometry. (D) Impaired NBS1 function does not induce cell death in macrophages treated with M-CSF or IFN-γ. Cells were treated as in (B), and viability was determined by using PI staining and flow cytometry. (E) The levels of Bcl2 mRNA expression were determined by qPCR in BMDMs treated with M-CSF or IFN-γ for the indicated times. Each experiment was performed in triplicate and the results are shown as mean ± SD. All assays are representative of at least 4 independent experiments showing similar results. **P < .01 and ***P < .001 in relation to the controls when all the independent experiments had been compared.

Figure 2

Reduced proliferation of macrophages from Nbs1∆B/∆B mice. (A) The number of total cells from the BM at day 0 of differentiation (BM after it was flushed from bones) and after 6 days of differentiation (in media containing M-CSF) from the indicated genotypes. (B) Macrophages were starved of growth factor for 18 hours, incubated for 1 hour with the indicated stimuli (LPS or IFN-γ), washed, and incubated for 24 hours with M-CSF (10 ng/mL) in a hyperoxic (20% O2) or hypoxic (1% O2) environment, and proliferation was quantified by incorporation of tritium. (C) Macrophages were starved of growth factor for 18 hours and incubated for 24 hours with M-CSF. Cell cycle distribution was determined by staining fixed cells with PI and assessment by flow cytometry. (D) Impaired NBS1 function does not induce cell death in macrophages treated with M-CSF or IFN-γ. Cells were treated as in (B), and viability was determined by using PI staining and flow cytometry. (E) The levels of Bcl2 mRNA expression were determined by qPCR in BMDMs treated with M-CSF or IFN-γ for the indicated times. Each experiment was performed in triplicate and the results are shown as mean ± SD. All assays are representative of at least 4 independent experiments showing similar results. **P < .01 and ***P < .001 in relation to the controls when all the independent experiments had been compared.

Close modal

Because M-CSF induced NBS1 protein levels (Figure 1B), we examined whether macrophage proliferation was affected in Nbs1∆B/∆B mice. Once differentiated after 7 days in M-CSF, BMDMs were plated and deprived of M-CSF for 16 to 18 hours. Cells were subsequently treated for 1 hour with the indicated stimuli, washed, and further incubated with M-CSF for 24 hours, and proliferation was measured by 3H-thymidine incorporation. Nbs1∆B/∆B macrophages exhibited a decreased level of M-CSF-dependent proliferative capacity (Figure 2B). To eliminate the possibility that these differences were caused by an increase in DNA damage as a result of atmospheric ROS, experiments were repeated in a hypoxic chamber (1% O2), and similar differences were observed between macrophages from WT and Nbs1∆B/∆B mice (Figure 2B).

Because macrophage proliferation was affected in Nbs1∆B/∆B cells, we next evaluated the cell cycle distribution. After cell cycle synchronization by 18 hours of M-CSF withdrawal, cells were cultured in an M-CSF-rich environment for 24 hours. Cells were subsequently fixed and the DNA content was assessed with PI staining. This analysis demonstrated that the amount of Nbs1∆B/∆B macrophages in S phase was reduced compared with WT macrophages in both hypoxic and hyperoxic conditions (Figure 2C and supplemental Figure 3), further corroborating the proliferative deficiency. This demonstrated that NBS1 was crucial for macrophage proliferation and cell cycle progression in vitro.

In addition to suppressing replication-associated DNA damage, the MRE11 complex is critical for apoptosis in various contexts, including oncogene activation and exogenous DNA damage.8,33-36  Therefore, we examined the impact of the Nbs1∆B allele on apoptosis in stimulated macrophages. Cells were incubated with M-CSF or IFN-γ for 24 hours, and sub-G1/G0 DNA was measured after PI staining. Sub-G1/G0 levels were very low and no significant differences were observed between WT and Nbs1∆B/∆B BMDMs (Figure 2D). The expression of Bcl2, a major proapoptotic gene, was also unaffected by genotype (Figure 2E). These results demonstrated that apoptosis was not affected by the Nbs1∆B/∆B allele in macrophages, consistent with previous results in lymphocytes and brain,35,36  and thus decreased proliferation was not a consequence of increased cell death.

Delayed differentiation in macrophages from Nbs1∆B/∆B mice

We then further examined the effect of the Nbs1∆B/∆B allele on macrophage differentiation. BMDMs were counted, and after 6 days in culture, they were stained with markers of macrophage differentiation, including CD11b, F4/80, and Ly6C. Macrophages from Nbs1∆B/∆B mice expressed lower levels of CD11b and F4/80 and higher levels of Ly6C (Figure 3A). The altered expression of these markers indicated impaired macrophage differentiation in Nbs1∆B/∆B mice. We examined the expression levels of these differentiation markers during in vitro macrophage differentiation. After 7 days, in the CD11b+ population of control WT cells, the expression of Ly6Chigh marker disappeared. However, in cells from Nbs1∆B/∆B mice, a significant difference in Ly6Chigh expression was observed after 7 days (Figure 3B), indicating that macrophages from Nbs1∆B/∆B mice have delayed maturation.

Figure 3

Impaired differentiation in macrophages from Nbs1∆B/∆B mice. Macrophages were differentiated from the BM of WT and Nbs1∆B/∆B mice, and the surface expression of differentiation markers was determined by using antibodies and flow cytometry. (A) After 6 days of differentiation, cells were stained. (B) Time course of differentiation of Ly6ChighCD11b+ and F4/80+CD11b+ cells. Each experiment was performed in triplicate and the results are shown as mean ± SD. All assays are representative of at least 4 independent experiments showing similar results. *P < .05, **P < .01, and ***P < .001 in relation to the controls when all the independent experiments had been compared.

Figure 3

Impaired differentiation in macrophages from Nbs1∆B/∆B mice. Macrophages were differentiated from the BM of WT and Nbs1∆B/∆B mice, and the surface expression of differentiation markers was determined by using antibodies and flow cytometry. (A) After 6 days of differentiation, cells were stained. (B) Time course of differentiation of Ly6ChighCD11b+ and F4/80+CD11b+ cells. Each experiment was performed in triplicate and the results are shown as mean ± SD. All assays are representative of at least 4 independent experiments showing similar results. *P < .05, **P < .01, and ***P < .001 in relation to the controls when all the independent experiments had been compared.

Close modal

Increased senescence in macrophages from Nbs1∆B/∆B mice

Cell cycle arrest and reduced proliferation, as well as cellular senescence, can be caused by DNA damage.37  To determine whether reduced proliferation of Nbs1∆B/∆B macrophages was a result of increased DNA damage, we examined senescence markers. We analyzed telomere length by using real-time PCR and found that it was, on average, shorter in Nbs1∆B/∆B macrophages (Figure 4A). In addition, Nbs1∆B/∆B macrophages exhibited higher levels of chromosomal aberrations when compared with control macrophages at day 7 (Figure 4B and supplemental Figure 4), and this correlated with higher ROS levels (Figure 4D), reminiscent of the results in Figure 1D, in which Nbs1∆B/∆B macrophages had increased γH2AX foci that were reduced upon treatment with N-acetyl-l-cysteine. Next, we assessed the mRNA levels of p21waf-1, which is induced by DNA damage and is considered a marker of senescence.38 p21waf-1 expression was determined by qPCR at different times after stimulation with either IFN-γ or M-CSF. In both cases, Nbs1∆B/∆B macrophages showed a higher level of p21waf-1 (Figure 4C), although it was induced with different kinetics. After stimulation with IFN-γ, p21waf-1 expression peaked at 3 hours, and after stimulation with M-CSF, expression peaked at 24 hours. Previously, we showed that senescent macrophages have lower MHC II expression.39  Consistent with the other indicators of senescence, we found that MHC II surface expression decreased in macrophages from Nbs1∆B/∆B mice (Figure 4E) after incubation of BMDMs with IFN-γ. Macrophages from Nbs1∆B/∆B mice treated with different proinflammatory cytokines showed increased expression of Il-6, Tnf-α and Ifn-β (Figure 4F). Collectively, these results suggested that increased DNA damage and impaired cell cycle progression in macrophages from Nbs1∆B/∆B mice may impair differentiation and promote the induction of senescence.

Figure 4

Increased senescence in macrophages from Nbs1∆B/∆B mice. (A) Shorter average telomere length in Nbs1∆B/∆B BMDMs. DNA from BMDMs was extracted and relative telomere length was determined by qPCR. (B) Metaphase spreads were performed as indicated in the “Materials and methods” section. Quantification of metaphase aberrations in 100 cells per mouse is shown (n = 3). (C) The expression of p21waf-1 was determined by qPCR from BMDMs stimulated with proinflammatory (IFN-γ) or proliferative (M-CSF) stimuli for the indicated times. (D) BMDMs were stimulated with M-CSF for 30 hours, and ROS levels were determined by flow cytometry using dichlorofluorescin diacetate (DCF-DA) as an indicator. (E) BMDM cells were activated with IFN- γ for 24 hours, and then the expression of MHC class II was determined by flow cytometry. (F) The macrophages from Nbs1∆B/∆B mice express higher levels of proinflammatory cytokines. BMDMs were incubated for 6 hours with the indicated stimuli, and the expression of Il-6, Ifn-β, and Tnf-α was determined by qPCR. Each point was performed in triplicate and the results are shown as mean ± SD. All assays are representative of at least 4 independent experiments showing similar results. *P < .05, **P < .01, and ***P < .001 in relation to the controls when all the independent experiments had been compared. MFI, mean fluorescent intensity.

Figure 4

Increased senescence in macrophages from Nbs1∆B/∆B mice. (A) Shorter average telomere length in Nbs1∆B/∆B BMDMs. DNA from BMDMs was extracted and relative telomere length was determined by qPCR. (B) Metaphase spreads were performed as indicated in the “Materials and methods” section. Quantification of metaphase aberrations in 100 cells per mouse is shown (n = 3). (C) The expression of p21waf-1 was determined by qPCR from BMDMs stimulated with proinflammatory (IFN-γ) or proliferative (M-CSF) stimuli for the indicated times. (D) BMDMs were stimulated with M-CSF for 30 hours, and ROS levels were determined by flow cytometry using dichlorofluorescin diacetate (DCF-DA) as an indicator. (E) BMDM cells were activated with IFN- γ for 24 hours, and then the expression of MHC class II was determined by flow cytometry. (F) The macrophages from Nbs1∆B/∆B mice express higher levels of proinflammatory cytokines. BMDMs were incubated for 6 hours with the indicated stimuli, and the expression of Il-6, Ifn-β, and Tnf-α was determined by qPCR. Each point was performed in triplicate and the results are shown as mean ± SD. All assays are representative of at least 4 independent experiments showing similar results. *P < .05, **P < .01, and ***P < .001 in relation to the controls when all the independent experiments had been compared. MFI, mean fluorescent intensity.

Close modal

Enhanced inflammatory responses in Nbs1∆B/∆B mice

During inflammation, the numbers and functional activities of immune cells are crucial to mount an effective response. To determine whether the deficiencies we observed in Nbs1∆B/∆B macrophages had an effect on the immune response in vivo, we used the DNFB ear irritation model.27  Although the DNFB ear irritation model is not a macrophage-specific model, macrophages incorporated from the bloodstream play a critical role in the early stages of this inflammation as well as in its resolution.17  Mice were treated on the right ear with DNFB and on the left ear with acetone (control) and were euthanized after 7 days. Ears were punched at the inflammatory loci, the punch was weighed, and tissue was collected for histology and RNA extraction. Ear thickness was measured after H&E staining, and levels of proinflammatory and anti-inflammatory genes were measured by qPCR. After treatment with DNFB, ears from Nbs1∆B/∆B mice were heavier and thicker than those of WT littermates (Figure 5A). The mRNA levels of proinflammatory and anti-inflammatory genes were also affected (Figure 5B). These results demonstrated that Nbs1∆B/∆B mice had an impaired immune response with an exacerbated proinflammatory response.

Figure 5

Exacerbated inflammation in Nbs1∆B/∆B mice treated with DNFB. (A) A representative sample of histologic images of DNFB-treated ears (at 7 day) is shown. At the right, the net weight of ears treated for 7 days with DNFB (8 mice) is shown. This was calculated by subtracting the weight of the DNFB-treated right ear from that of the acetone-treated left ear that was used as control. The ear thickness measurements were taken from the histologic images using Fiji software. Only DNFB-treated ear thickness is shown. AU, arbitrary unit. (B) Ear sample-derived mRNA was used to determine the expression of proinflammatory and anti-inflammatory markers by qPCR. The results are shown as individual values and as the mean of the 8 WT and 8 Nbs1∆B/∆B mice. *P < .05 and **P < .01 in relation to the corresponding controls.

Figure 5

Exacerbated inflammation in Nbs1∆B/∆B mice treated with DNFB. (A) A representative sample of histologic images of DNFB-treated ears (at 7 day) is shown. At the right, the net weight of ears treated for 7 days with DNFB (8 mice) is shown. This was calculated by subtracting the weight of the DNFB-treated right ear from that of the acetone-treated left ear that was used as control. The ear thickness measurements were taken from the histologic images using Fiji software. Only DNFB-treated ear thickness is shown. AU, arbitrary unit. (B) Ear sample-derived mRNA was used to determine the expression of proinflammatory and anti-inflammatory markers by qPCR. The results are shown as individual values and as the mean of the 8 WT and 8 Nbs1∆B/∆B mice. *P < .05 and **P < .01 in relation to the corresponding controls.

Close modal

To examine the role of NBS1 in another in vivo model of inflammation, we used the intraperitoneal injection of zymosan, yeast fragments that bind TLR2, inducing peritonitis and triggering a rapid response of the immune system. At 24 hours to 60 hours after zymosan injection, macrophages are the most abundant cells accumulating at the peritoneal cavity.40  This is due to not only inflammatory macrophage recruitment but also to local macrophage proliferation that is M-CSF dependent.41  We injected zymosan into the peritoneal cavity of both WT and Nbs1∆B/∆B mice, and after 48 hours, we euthanized the mice and performed peritoneal lavages. The cells obtained were stained for CD11b, F4/80, and the intracellular proliferation marker Ki-67. By using flow cytometry, we differentiated proliferating macrophages from other proliferating cell types (Figure 6A). We injected PBS as a control, which did not affect the total number of proliferating cells or numbers of proliferating macrophages in WT or Nbs1∆B/∆B mice (Figure 6B). However, when we induced a sterile inflammation by zymosan injection, the percentage of macrophages was drastically decreased in the peritoneal cavity of Nbs1∆B/∆B mice (Figure 6B). Under these experimental conditions, we were unable to determine whether the cells that proliferate are of BM origin (moving from the blood) or from the peritoneal cavity. However, the analysis of peritoneal cells in vitro showed decreased proliferation in cells from Nbs1∆B/∆B mice (Figure 6C). Therefore, both BMDMs and peritoneal macrophage populations derived from Nbs1∆B/∆B mice showed impaired proliferative capacity.

Figure 6

Reduced macrophage proliferation from Nbs1∆B/∆B mice in a model of sterile inflammation. Control and Nbs1∆B/∆B mice were injected in the peritoneal cavity with 2 × 106 zymosan particles or PBS (control). (A) Gating strategy of peritoneal cells after staining with anti-CD11b, anti-F4/80, and anti-Ki67 (cell marker for proliferation). FS-W, forward scatter–width; FSS, forward scatter signal; SS-A, side scatter–area. (B) The percentage of Ki-67+F4/80+ cells (macrophages) and Ki-67+F4/80 cells was determined for animals treated with either PBS or zymosan. For the control experiments, 7 control mice and 6 Nbs1∆B mice were used, and for the experiment with zymosan, 8 control mice and 7 Nbs1∆B/∆B mice were used. ***P < .001. (C) Proliferation of peritoneal macrophages. Macrophages were obtained by peritoneal lavage and purified by adhesion. Proliferation was determined by thymidine incorporation after 24 hours of incubation with M-CSF (n = 4). cpm, counts per minute. The results are shown as mean ± SD. *P < .01 in relation to the corresponding controls.

Figure 6

Reduced macrophage proliferation from Nbs1∆B/∆B mice in a model of sterile inflammation. Control and Nbs1∆B/∆B mice were injected in the peritoneal cavity with 2 × 106 zymosan particles or PBS (control). (A) Gating strategy of peritoneal cells after staining with anti-CD11b, anti-F4/80, and anti-Ki67 (cell marker for proliferation). FS-W, forward scatter–width; FSS, forward scatter signal; SS-A, side scatter–area. (B) The percentage of Ki-67+F4/80+ cells (macrophages) and Ki-67+F4/80 cells was determined for animals treated with either PBS or zymosan. For the control experiments, 7 control mice and 6 Nbs1∆B mice were used, and for the experiment with zymosan, 8 control mice and 7 Nbs1∆B/∆B mice were used. ***P < .001. (C) Proliferation of peritoneal macrophages. Macrophages were obtained by peritoneal lavage and purified by adhesion. Proliferation was determined by thymidine incorporation after 24 hours of incubation with M-CSF (n = 4). cpm, counts per minute. The results are shown as mean ± SD. *P < .01 in relation to the corresponding controls.

Close modal

After proinflammatory stimulation, macrophages produce increased ROS that are able to extensively damage DNA.42-44  Under these stimuli (IFN-γ or LPS), we detected increased levels of NBS1. In Nbs1∆B/∆B macrophages, this stimulation led to increased DNA damage that could be strongly reduced by the addition of N-acetyl-l-cysteine, indicating that the MRE11 complex was important for suppressing ROS-induced DNA damage (Figure 1D). However, even in the absence of ROS-inducing stimuli, the basal levels of DNA breaks were higher in Nbs1∆B/∆B macrophages, potentially reflecting defects in DNA surveillance during the rapid proliferation that precedes differentiation. Consistent with this possibility, stimulation with M-CSF that leads to high levels of proliferation was accompanied by increased levels of chromatid breaks in Nbs1∆B/∆B macrophages (Figure 4B and supplemental Figure 2). The increased NBS1 protein levels observed in activated macrophages were the result of posttranscriptional regulation because we did not observe differences in mRNA levels after any of the treatments (Figure 1A). Because little is known regarding the regulation of NBS1 protein levels, further studies will be required to elucidate the mechanisms by which NBS1 levels are controlled in macrophages. We believe they are likely to be somewhat specific, because no differences were observed in the levels of other DDR proteins, including ATR, CHK2, and KU80, after stimulation. Together, our results suggest that the MRE11 complex is crucial for preventing DNA damage in macrophages during replication and after proinflammatory activation.

In mice, the complete deficiency of Nbs1 led to embryonic lethality.7  By using a mouse model expressing a hypomorphic allele of Nbs120  that does not compromise animal viability or accelerate mortality, we confirmed that NBS1 function was important for macrophage function because Nbs1∆B/∆B macrophages showed impaired proliferation and delayed differentiation. This was accompanied by the induction of 2 of the classical markers associated with aging in macrophages, increased numbers of chromosomal aberrations and telomere shortening,24  as well as a lack of MHC II induction by IFN-γ39  and increased levels of p21, a marker of senescence. Given the roles of the MRE11 complex in DNA replication, cell cycle control, and telomere maintenance,7,45  we believe that this premature aging of macrophages is the most likely explanation for the reduced proliferation and delayed differentiation, because we did not observe changes in the induction of apoptosis, suggesting that cell cycle abnormalities were not the result of increased cell death.

Our in vitro observations suggested that the accumulation of increased DNA damage may compromise macrophage function and lead to premature attrition in vivo. Monocytes are more prone to DNA damage than dendritic cells and macrophages, exhibiting increased apoptosis upon ROS exposure resulting from impaired DSB repair.46  Because Nbs1∆B/∆B mice have defective DSB responses, it is possible that over time, DNA damage is accumulated in monocytes and macrophages.

Consistent with our observations in vitro, we also observed an enhanced inflammatory response in Nbs1∆B/∆B mice after treatment with DNFB. Persistent DNA damage has been linked to senescence and has been shown to trigger inflammatory cytokines, as we have observed in Nbs1∆B/∆B macrophages. This response has been termed the “senescence-associated secretory phenotype” or SASP, and its function has been linked to a variety of age-related pathological outcomes, including tumorigenesis and sterile chronic inflammation.47,48  Recent work has demonstrated that the MRE11 complex and ATM are critical for the innate immune response to cytosolic DNA in bone marrow-derived dendritic cells (BMDCs)49,50  and in BMDMs.51  In response to cytosolic DNA generated by increased DNA damage or polyinosinic-polycytidylic acid, BMDCs activate the STING pathway to trigger SASP-inducing inflammatory pathways mediated by IL-1β and the inflammosome.49,50  In this context, cytosolic DNA sensing has been attributed to ATM and interactions between RAD50 and the CARD9 protein.49  The Nbs1∆B allele leads to mislocalization of some of the MRE11-RAD50 pool to the cytoplasm and therefore may lead to both increased cytosolic DNA and enhanced signaling in BMDCs, because NBS1 is not strictly required for the cytosolic responses.49 

In addition to the MRE11 complex and ATM, a number of additional cytosolic nucleic acid sensors have been identified and are associated with the suppression of type I IFN production. This includes Trex1, which is mutated in systemic lupus erythematosus and implicated in the DNA damage response.52,53  We recently reported enhanced proinflammatory activation in macrophages from mice lacking Trex1,22  and the release of cytokines has been attributed to phenotypic variations in patients with Aicardi-Goutières syndrome.54  Mutations that lead to activation of innate immunity by inducing the secretion of type I IFN collectively may represent “interferonopathies”,55  disorders that are characterized by neurologic and dermatologic features provoked by autoimmunity.

The SASP likely shares many common features with the interferonopathies. Both may originate from the malfunction of proteins involved in responding to DNA damage that can trigger large amounts of both type I IFNs and inflammatory cytokines, and their co-occurrence may be important to consider in human disease. These findings suggest the relationship between the SASP and IFN response and the role of the DNA damage response should be examined further in different types of immune cells.

In conclusion, our results demonstrate that NBS1 plays an important role in macrophage differentiation, proliferation, and inflammatory responses in vivo. Here we have described what is, to the best of our knowledge, the first functional macrophage defects resulting from mutations in Nbs1. Because NBS patients present with immunodeficiency, which is a major health threat to these patients, a complete understanding of the role of the MRE11 complex in immune system development and homeostasis is essential and the role of macrophages in NBS pathology should be further considered.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors acknowledge Gemma Lopez, Natalia Plana, Lluis Palenzuela, and Jaume Comas for their excellent technical assistance and Andre Nussenzweig and Elsa Callen for reagents.

This work was supported by Formación del Profesorado Universitario grants AP2010-5396 (S.P.-L.) and AP2012-02327 (J.T.) from the Ministerio de Educación, Cultura y Deporte, by Formación del Personal Investigador grant BES-2012-055074 (J.A.C.-S.) from the Ministerio de Economía y Competitividad (MINECO), and by grants BFU2012-39521 (T.H.S.) and BFU2011-23662 (A.C.) from the MINECO.

Contribution: S.P.-L. designed and performed research, analyzed data, and wrote the paper; J.T. and J.A.C.-S. performed research; J.L. designed the research; and T.H.S. and A.C. designed the research, analyzed data, and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Antonio Celada, Parc Cientific of Barcelona, Baldiri Reixac 10, 08028 Barcelona, Spain; e-mail: acelada@ub.edu.

1
Jackson
 
SP
Bartek
 
J
The DNA-damage response in human biology and disease.
Nature
2009
, vol. 
461
 
7267
(pg. 
1071
-
1078
)
2
Bohgaki
 
T
Bohgaki
 
M
Hakem
 
R
DNA double-strand break signaling and human disorders.
Genome Integr
2010
, vol. 
1
 
1
pg. 
15
 
3
Chapman
 
JR
Taylor
 
MR
Boulton
 
SJ
Playing the end game: DNA double-strand break repair pathway choice.
Mol Cell
2012
, vol. 
47
 
4
(pg. 
497
-
510
)
4
Stracker
 
TH
Roig
 
I
Knobel
 
PA
Marjanović
 
M
 
The ATM signaling network in development and disease. Front Genet. 2013;4:37
5
Stracker
 
TH
Petrini
 
JH
Working together and apart: the twisted relationship of the Mre11 complex and Chk2 in apoptosis and tumor suppression.
Cell Cycle
2008
, vol. 
7
 
23
(pg. 
3618
-
3621
)
6
Rein
 
K
Stracker
 
TH
The MRE11 complex: an important source of stress relief.
Exp Cell Res
2014
, vol. 
329
 
1
(pg. 
162
-
169
)
7
Stracker
 
TH
Petrini
 
JH
The MRE11 complex: starting from the ends.
Nat Rev Mol Cell Biol
2011
, vol. 
12
 
2
(pg. 
90
-
103
)
8
Difilippantonio
 
S
Celeste
 
A
Kruhlak
 
MJ
, et al. 
Distinct domains in Nbs1 regulate irradiation-induced checkpoints and apoptosis.
J Exp Med
2007
, vol. 
204
 
5
(pg. 
1003
-
1011
)
9
Varon
 
R
Vissinga
 
C
Platzer
 
M
, et al. 
Nibrin, a novel DNA double-strand break repair protein, is mutated in Nijmegen breakage syndrome.
Cell
1998
, vol. 
93
 
3
(pg. 
467
-
476
)
10
Carney
 
JP
Maser
 
RS
Olivares
 
H
, et al. 
The hMre11/hRad50 protein complex and Nijmegen breakage syndrome: linkage of double-strand break repair to the cellular DNA damage response.
Cell
1998
, vol. 
93
 
3
(pg. 
477
-
486
)
11
Varon
 
R
Seemanova
 
E
Chrzanowska
 
K
, et al. 
Clinical ascertainment of Nijmegen breakage syndrome (NBS) and prevalence of the major mutation, 657del5, in three Slav populations.
Eur J Hum Genet
2000
, vol. 
8
 
11
(pg. 
900
-
902
)
12
van der Burg
 
M
Pac
 
M
Berkowska
 
MA
, et al. 
Loss of juxtaposition of RAG-induced immunoglobulin DNA ends is implicated in the precursor B-cell differentiation defect in NBS patients.
Blood
2010
, vol. 
115
 
23
(pg. 
4770
-
4777
)
13
Piątosa
 
B
van der Burg
 
M
Siewiera
 
K
, et al. 
The defect in humoral immunity in patients with Nijmegen breakage syndrome is explained by defects in peripheral B lymphocyte maturation.
Cytometry A
2012
, vol. 
81
 
10
(pg. 
835
-
842
)
14
Xaus
 
J
Comalada
 
M
Valledor
 
AF
, et al. 
Molecular mechanisms involved in macrophage survival, proliferation, activation or apoptosis.
Immunobiology
2001
, vol. 
204
 
5
(pg. 
543
-
550
)
15
Barreda
 
DR
Hanington
 
PC
Belosevic
 
M
Regulation of myeloid development and function by colony stimulating factors.
Dev Comp Immunol
2004
, vol. 
28
 
5
(pg. 
509
-
554
)
16
Sica
 
A
Mantovani
 
A
Macrophage plasticity and polarization: in vivo veritas.
J Clin Invest
2012
, vol. 
122
 
3
(pg. 
787
-
795
)
17
Guilliams
 
M
Ginhoux
 
F
Jakubzick
 
C
, et al. 
Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny.
Nat Rev Immunol
2014
, vol. 
14
 
8
(pg. 
571
-
578
)
18
Slupphaug
 
G
Kavli
 
B
Krokan
 
HE
The interacting pathways for prevention and repair of oxidative DNA damage.
Mutat Res
2003
, vol. 
531
 
1-2
(pg. 
231
-
251
)
19
Valledor
 
AF
Comalada
 
M
Santamaría-Babi
 
LF
Lloberas
 
J
Celada
 
A
Macrophage proinflammatory activation and deactivation: a question of balance.
Adv Immunol
2010
, vol. 
108
 (pg. 
1
-
20
)
20
Williams
 
BR
Mirzoeva
 
OK
Morgan
 
WF
Lin
 
J
Dunnick
 
W
Petrini
 
JH
A murine model of Nijmegen breakage syndrome.
Curr Biol
2002
, vol. 
12
 
8
(pg. 
648
-
653
)
21
Celada
 
A
Borràs
 
FE
Soler
 
C
, et al. 
The transcription factor PU.1 is involved in macrophage proliferation.
J Exp Med
1996
, vol. 
184
 
1
(pg. 
61
-
69
)
22
Pereira-Lopes
 
S
Celhar
 
T
Sans-Fons
 
G
, et al. 
The exonuclease Trex1 restrains macrophage proinflammatory activation.
J Immunol
2013
, vol. 
191
 
12
(pg. 
6128
-
6135
)
23
Xaus
 
J
Cardó
 
M
Valledor
 
AF
Soler
 
C
Lloberas
 
J
Celada
 
A
Interferon gamma induces the expression of p21waf-1 and arrests macrophage cell cycle, preventing induction of apoptosis.
Immunity
1999
, vol. 
11
 
1
(pg. 
103
-
113
)
24
Sebastián
 
C
Herrero
 
C
Serra
 
M
Lloberas
 
J
Blasco
 
MA
Celada
 
A
Telomere shortening and oxidative stress in aged macrophages results in impaired STAT5a phosphorylation.
J Immunol
2009
, vol. 
183
 
4
(pg. 
2356
-
2364
)
25
Knuutila
 
S
Pohjanpelto
 
P
Polyamine starvation causes parallel increase in nuclear and chromosomal aberrations in a polyamine-dependent strain of CHO.
Exp Cell Res
1983
, vol. 
145
 
1
(pg. 
222
-
226
)
26
Cash
 
JL
White
 
GE
Greaves
 
DR
Chapter 17. Zymosan-induced peritonitis as a simple experimental system for the study of inflammation.
Methods Enzymol
2009
, vol. 
461
 (pg. 
379
-
396
)
27
Bonneville
 
M
Chavagnac
 
C
Vocanson
 
M
, et al. 
Skin contact irritation conditions the development and severity of allergic contact dermatitis.
J Invest Dermatol
2007
, vol. 
127
 
6
(pg. 
1430
-
1435
)
28
Comalada
 
M
Xaus
 
J
Sánchez
 
E
Valledor
 
AF
Celada
 
A
Macrophage colony-stimulating factor-, granulocyte-macrophage colony-stimulating factor-, or IL-3-dependent survival of macrophages, but not proliferation, requires the expression of p21(Waf1) through the phosphatidylinositol 3-kinase/Akt pathway.
Eur J Immunol
2004
, vol. 
34
 
8
(pg. 
2257
-
2267
)
29
Collins
 
MK
Marvel
 
J
Malde
 
P
Lopez-Rivas
 
A
Interleukin 3 protects murine bone marrow cells from apoptosis induced by DNA damaging agents.
J Exp Med
1992
, vol. 
176
 
4
(pg. 
1043
-
1051
)
30
Yeramian
 
A
Martin
 
L
Serrat
 
N
, et al. 
Arginine transport via cationic amino acid transporter 2 plays a critical regulatory role in classical or alternative activation of macrophages.
J Immunol
2006
, vol. 
176
 
10
(pg. 
5918
-
5924
)
31
Medzhitov
 
R
Toll-like receptors and innate immunity.
Nat Rev Immunol
2001
, vol. 
1
 
2
(pg. 
135
-
145
)
32
Costanzo
 
V
Robertson
 
K
Bibikova
 
M
, et al. 
Mre11 protein complex prevents double-strand break accumulation during chromosomal DNA replication.
Mol Cell
2001
, vol. 
8
 
1
(pg. 
137
-
147
)
33
Rogoff
 
HA
Pickering
 
MT
Frame
 
FM
, et al. 
Apoptosis associated with deregulated E2F activity is dependent on E2F1 and Atm/Nbs1/Chk2.
Mol Cell Biol
2004
, vol. 
24
 
7
(pg. 
2968
-
2977
)
34
Wang
 
H
Zhang
 
X
Teng
 
L
Legerski
 
RJ
DNA damage checkpoint recovery and cancer development.
Exp Cell Res
2015
, vol. 
334
 
2
(pg. 
350
-
358
)
35
Stracker
 
TH
Couto
 
SS
Cordon-Cardo
 
C
Matos
 
T
Petrini
 
JH
Chk2 suppresses the oncogenic potential of DNA replication-associated DNA damage.
Mol Cell
2008
, vol. 
31
 
1
(pg. 
21
-
32
)
36
Shull
 
ER
Lee
 
Y
Nakane
 
H
, et al. 
Differential DNA damage signaling accounts for distinct neural apoptotic responses in ATLD and NBS.
Genes Dev
2009
, vol. 
23
 
2
(pg. 
171
-
180
)
37
Aguilera
 
A
García-Muse
 
T
Causes of genome instability.
Annu Rev Genet
2013
, vol. 
47
 (pg. 
1
-
32
)
38
Sperka
 
T
Wang
 
J
Rudolph
 
KL
DNA damage checkpoints in stem cells, ageing and cancer.
Nat Rev Mol Cell Biol
2012
, vol. 
13
 
9
(pg. 
579
-
590
)
39
Herrero
 
C
Marqués
 
L
Lloberas
 
J
Celada
 
A
IFN-gamma-dependent transcription of MHC class II IA is impaired in macrophages from aged mice.
J Clin Invest
2001
, vol. 
107
 
4
(pg. 
485
-
493
)
40
Choi
 
H
Lee
 
RH
Bazhanov
 
N
Oh
 
JY
Prockop
 
DJ
Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-κB signaling in resident macrophages.
Blood
2011
, vol. 
118
 
2
(pg. 
330
-
338
)
41
Davies
 
LC
Rosas
 
M
Jenkins
 
SJ
, et al. 
Distinct bone marrow-derived and tissue-resident macrophage lineages proliferate at key stages during inflammation.
Nat Commun
2013
, vol. 
4
 pg. 
1886
 
42
Forman
 
HJ
Torres
 
M
Redox signaling in macrophages.
Mol Aspects Med
2001
, vol. 
22
 
4-5
(pg. 
189
-
216
)
43
Berquist
 
BR
Wilson
 
DM
Pathways for repairing and tolerating the spectrum of oxidative DNA lesions.
Cancer Lett
2012
, vol. 
327
 
1-2
(pg. 
61
-
72
)
44
Gwinn
 
MR
Vallyathan
 
V
Respiratory burst: role in signal transduction in alveolar macrophages.
J Toxicol Environ Health B Crit Rev
2006
, vol. 
9
 
1
(pg. 
27
-
39
)
45
Deng
 
Y
Guo
 
X
Ferguson
 
DO
Chang
 
S
Multiple roles for MRE11 at uncapped telomeres.
Nature
2009
, vol. 
460
 
7257
(pg. 
914
-
918
)
46
Bauer
 
M
Goldstein
 
M
Christmann
 
M
Becker
 
H
Heylmann
 
D
Kaina
 
B
Human monocytes are severely impaired in base and DNA double-strand break repair that renders them vulnerable to oxidative stress.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
52
(pg. 
21105
-
21110
)
47
Rodier
 
F
Coppé
 
JP
Patil
 
CK
, et al. 
Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion.
Nat Cell Biol
2009
, vol. 
11
 
8
(pg. 
973
-
979
)
48
Lasry
 
A
Ben-Neriah
 
Y
Senescence-associated inflammatory responses: aging and cancer perspectives.
Trends Immunol
2015
, vol. 
36
 
4
(pg. 
217
-
228
)
49
Roth
 
S
Rottach
 
A
Lotz-Havla
 
AS
, et al. 
Rad50-CARD9 interactions link cytosolic DNA sensing to IL-1β production.
Nat Immunol
2014
, vol. 
15
 
6
(pg. 
538
-
545
)
50
Kondo
 
T
Kobayashi
 
J
Saitoh
 
T
, et al. 
DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA and induces type I interferon by regulating STING trafficking.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
8
(pg. 
2969
-
2974
)
51
Härtlova
 
A
Erttmann
 
SF
Raffi
 
FA
, et al. 
DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity.
Immunity
2015
, vol. 
42
 
2
(pg. 
332
-
343
)
52
Rice
 
GI
Rodero
 
MP
Crow
 
YJ
Human disease phenotypes associated with mutations in TREX1.
J Clin Immunol
2015
, vol. 
35
 
3
(pg. 
235
-
243
)
53
Yang
 
YG
Lindahl
 
T
Barnes
 
DE
Trex1 exonuclease degrades ssDNA to prevent chronic checkpoint activation and autoimmune disease.
Cell
2007
, vol. 
131
 
5
(pg. 
873
-
886
)
54
Cuadrado
 
E
Michailidou
 
I
van Bodegraven
 
EJ
, et al. 
Phenotypic variation in Aicardi-Goutières syndrome explained by cell-specific IFN-stimulated gene response and cytokine release.
J Immunol
2015
, vol. 
194
 
8
(pg. 
3623
-
3633
)
55
Crow
 
YJ
Manel
 
N
Aicardi-Goutières syndrome and the type I interferonopathies.
Nat Rev Immunol
2015
, vol. 
15
 
7
(pg. 
429
-
440
)
Sign in via your Institution