Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

AIM2 inflammasome surveillance of DNA damage shapes neurodevelopment

Subjects

Abstract

Neurodevelopment is characterized by rapid rates of neural cell proliferation and differentiation followed by massive cell death in which more than half of all recently generated brain cells are pruned back. Large amounts of DNA damage, cellular debris, and by-products of cellular stress are generated during these neurodevelopmental events, all of which can potentially activate immune signalling. How the immune response to this collateral damage influences brain maturation and function remains unknown. Here we show that the AIM2 inflammasome contributes to normal brain development and that disruption of this immune sensor of genotoxic stress leads to behavioural abnormalities. During infection, activation of the AIM2 inflammasome in response to double-stranded DNA damage triggers the production of cytokines as well as a gasdermin-D-mediated form of cell death known as pyroptosis1,2,3,4. We observe pronounced AIM2 inflammasome activation in neurodevelopment and find that defects in this sensor of DNA damage result in anxiety-related behaviours in mice. Furthermore, we show that the AIM2 inflammasome contributes to central nervous system (CNS) homeostasis specifically through its regulation of gasdermin-D, and not via its involvement in the production of the cytokines IL-1 and/or IL-18. Consistent with a role for this sensor of genomic stress in the purging of genetically compromised CNS cells, we find that defective AIM2 inflammasome signalling results in decreased neural cell death both in response to DNA damage-inducing agents and during neurodevelopment. Moreover, mutations in AIM2 lead to excessive accumulation of DNA damage in neurons as well as an increase in the number of neurons that incorporate into the adult brain. Our findings identify the inflammasome as a crucial player in establishing a properly formed CNS through its role in the removal of genetically compromised cells.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Inflammasome activation occurs in the CNS during neurodevelopment and disruption of the AIM2 inflammasome results in anxiety-related behaviours.
Fig. 2: Lack of gasdermin-D activation drives anxiety-like behaviours in mice.
Fig. 3: Activation of the AIM2 inflammasome in response to DNA damage coordinates CNS cell death and limits the accumulation of DNA damage in the brain.
Fig. 4: CNS-specific deletion of caspase-1 results in anxiety-like behaviours and DNA damage accumulation in the brain.

Similar content being viewed by others

Data availability

Source Data for Figs. 14 and Extended Data Figs. 1, 37, 9, 10 containing raw data for all experiments are provided with the paper. All other data are available from the corresponding author upon request.

References

  1. Fernandes-Alnemri, T., Yu, J. W., Datta, P., Wu, J. & Alnemri, E. S. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 458, 509–513 (2009).

    Article  ADS  CAS  Google Scholar 

  2. Hornung, V. et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458, 514–518 (2009).

    Article  ADS  CAS  Google Scholar 

  3. Rathinam, V. A. et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat. Immunol. 11, 395–402 (2010).

    Article  CAS  Google Scholar 

  4. Guo, H., Callaway, J. B. & Ting, J. P. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat. Med. 21, 677–687 (2015).

    Article  Google Scholar 

  5. Choi, G. B. et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 351, 933–939 (2016).

    Article  ADS  CAS  Google Scholar 

  6. Allan, S. M., Tyrrell, P. J. & Rothwell, N. J. Interleukin-1 and neuronal injury. Nat. Rev. Immunol. 5, 629–640 (2005).

    Article  CAS  Google Scholar 

  7. Felderhoff-Mueser, U., Schmidt, O. I., Oberholzer, A., Bührer, C. & Stahel, P. F. IL-18: a key player in neuroinflammation and neurodegeneration? Trends Neurosci. 28, 487–493 (2005).

    Article  CAS  Google Scholar 

  8. Dantzer, R., O’Connor, J. C., Freund, G. G., Johnson, R. W. & Kelley, K. W. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat. Rev. Neurosci. 9, 46–56 (2008).

    Article  CAS  Google Scholar 

  9. Garber, C. et al. Astrocytes decrease adult neurogenesis during virus-induced memory dysfunction via IL-1. Nat. Immunol. 19, 151–161 (2018).

    Article  CAS  Google Scholar 

  10. Walsh, J. G., Muruve, D. A. & Power, C. Inflammasomes in the CNS. Nat. Rev. Neurosci. 15, 84–97 (2014).

    Article  CAS  Google Scholar 

  11. Yamaguchi, Y. & Miura, M. Programmed cell death in neurodevelopment. Dev. Cell 32, 478–490 (2015).

    Article  CAS  Google Scholar 

  12. Kuan, C. Y. et al. The Jnk1 and Jnk2 protein kinases are required for regional specific apoptosis during early brain development. Neuron 22, 667–676 (1999).

    Article  CAS  Google Scholar 

  13. Cecconi, F., Alvarez-Bolado, G., Meyer, B. I., Roth, K. A. & Gruss, P. Apaf1 (CED-4 homolog) regulates programmed cell death in mammalian development. Cell 94, 727–737 (1998).

    Article  CAS  Google Scholar 

  14. Yoshida, H. et al. Apaf1 is required for mitochondrial pathways of apoptosis and brain development. Cell 94, 739–750 (1998).

    Article  CAS  Google Scholar 

  15. Tzeng, T. C. et al. A fluorescent reporter mouse for inflammasome assembly demonstrates an important role for cell-bound and free ASC specks during in vivo infection. Cell Rep. 16, 571–582 (2016).

    Article  CAS  Google Scholar 

  16. Herzog, K. H., Chong, M. J., Kapsetaki, M., Morgan, J. I. & McKinnon, P. J. Requirement for Atm in ionizing radiation-induced cell death in the developing central nervous system. Science 280, 1089–1091 (1998).

    Article  ADS  CAS  Google Scholar 

  17. Takashima, H. et al. Mutation of TDP1, encoding a topoisomerase I-dependent DNA damage repair enzyme, in spinocerebellar ataxia with axonal neuropathy. Nat. Genet. 32, 267–272 (2002).

    Article  CAS  Google Scholar 

  18. McKinnon, P. J. Maintaining genome stability in the nervous system. Nat. Neurosci. 16, 1523–1529 (2013).

    Article  CAS  Google Scholar 

  19. McKinnon, P. J. Genome integrity and disease prevention in the nervous system. Genes Dev. 31, 1180–1194 (2017).

    Article  CAS  Google Scholar 

  20. Schumacher, B., Garinis, G. A. & Hoeijmakers, J. H. Age to survive: DNA damage and aging. Trends Genet. 24, 77–85 (2008).

    Article  CAS  Google Scholar 

  21. Hoeijmakers, J. H. DNA damage, aging, and cancer. N. Engl. J. Med. 361, 1475–1485 (2009).

    Article  CAS  Google Scholar 

  22. Hu, B. et al. The DNA-sensing AIM2 inflammasome controls radiation-induced cell death and tissue injury. Science 354, 765–768 (2016).

    Article  ADS  CAS  Google Scholar 

  23. Denes, A. et al. AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain injury independently of NLRP3. Proc. Natl Acad. Sci. USA 112, 4050–4055 (2015).

    Article  ADS  CAS  Google Scholar 

  24. Adamczak, S. E. et al. Pyroptotic neuronal cell death mediated by the AIM2 inflammasome. J. Cereb. Blood Flow Metab. 34, 621–629 (2014).

    Article  CAS  Google Scholar 

  25. Wu, P. J., Liu, H. Y., Huang, T. N. & Hsueh, Y. P. AIM 2 inflammasomes regulate neuronal morphology and influence anxiety and memory in mice. Sci. Rep. 6, 32405 (2016).

    Article  ADS  CAS  Google Scholar 

  26. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

    Article  ADS  CAS  Google Scholar 

  27. Vanden Berghe, T., Linkermann, A., Jouan-Lanhouet, S., Walczak, H. & Vandenabeele, P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 15, 135–147 (2014).

    Article  CAS  Google Scholar 

  28. Filiano, A. J., Gadani, S. P. & Kipnis, J. How and why do T cells and their derived cytokines affect the injured and healthy brain? Nat. Rev. Neurosci. 18, 375–384 (2017).

    Article  CAS  Google Scholar 

  29. Hammond, T. R., Robinton, D. & Stevens, B. Microglia and the brain: complementary partners in development and disease. Annu. Rev. Cell Dev. Biol. 34, 523–544 (2018).

    Article  CAS  Google Scholar 

  30. Subba Rao, K. Mechanisms of disease: DNA repair defects and neurological disease. Nat. Clin. Pract. Neurol. 3, 162–172 (2007).

    Article  Google Scholar 

  31. Kuida, K. et al. Altered cytokine export and apoptosis in mice deficient in interleukin-1 beta converting enzyme. Science 267, 2000–2003 (1995).

    Article  ADS  CAS  Google Scholar 

  32. Kovarova, M. et al. NLRP1-dependent pyroptosis leads to acute lung injury and morbidity in mice. J. Immunol. 189, 2006–2016 (2012).

    Article  CAS  Google Scholar 

  33. Hou, B., Reizis, B. & DeFranco, A. L. Toll-like receptors activate innate and adaptive immunity by using dendritic cell-intrinsic and -extrinsic mechanisms. Immunity 29, 272–282 (2008).

    Article  CAS  Google Scholar 

  34. Glaccum, M. B. et al. Phenotypic and functional characterization of mice that lack the type I receptor for IL-1. J. Immunol. 159, 3364–3371 (1997).

    CAS  PubMed  Google Scholar 

  35. Hoshino, K. et al. Cutting edge: generation of IL-18 receptor-deficient mice: evidence for IL-1 receptor-related protein as an essential IL-18 binding receptor. J. Immunol. 162, 5041–5044 (1999).

    CAS  PubMed  Google Scholar 

  36. Wang, S. et al. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell 92, 501–509 (1998).

    Article  CAS  Google Scholar 

  37. Case, C. L. et al. Caspase-11 stimulates rapid flagellin-independent pyroptosis in response to Legionella pneumophila. Proc. Natl Acad. Sci. USA 110, 1851–1856 (2013).

    Article  ADS  CAS  Google Scholar 

  38. Tronche, F. et al. Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat. Genet. 23, 99–103 (1999).

    Article  CAS  Google Scholar 

  39. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  Google Scholar 

  40. Chen, S. H., Oyarzabal, E. A. & Hong, J. S. Preparation of rodent primary cultures for neuron-glia, mixed glia, enriched microglia, and reconstituted cultures with microglia. Methods Mol. Biol. 1041, 231–240 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Lukens laboratory and the Center for Brain Immunology and Glia (BIG) for discussions. This work was supported by The Hartwell Foundation (Individual Biomedical Research Award to J.R.L.), a Rettsyndrome.org grant (22349 to J.R.L.), The Owens Family Foundation (awarded to J.R.L.), and a NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation (27515 to J.R.L.). C.R.L was supported by a NIH National Institute of General Medical Sciences predoctoral training grant (3T32GM008328) and a Wagner Fellowship. A.C.B. was supported by a Medical Scientist Training Program Grant (5T32GM007267-38) and an Immunology Training Grant (5T32AI007496-25). H.E.E. was supported by a Cell and Molecular Biology Training Grant (T32GM008136). E.L.F. was supported by a National Multiple Sclerosis Foundation Postdoctoral Fellowship (FG-1707-28590). C.E.B. was supported by Hutcheson and Stull Undergraduate Research Fellowships.

Author information

Authors and Affiliations

Authors

Contributions

C.R.L. and J.R.L. designed the study; C.R.L., E.L.F., C.E.B., A.C.B., C.A.M., M.E.H., M.J.P., H.E.E. and J.R.L. performed experiments; C.R.L. and J.R.L. analysed data and wrote the manuscript; J.R.L. oversaw the project.

Corresponding author

Correspondence to John R. Lukens.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Molecular components of the AIM2 inflammasome are abundantly expressed in the brain during neurodevelopment.

ac, Brains from P5 (a) and P21 (b) wild-type mice were evaluated for the mRNA expression of inflammasome component genes (red) Casp1 (P5 n = 4, P21 n = 2), Aim2 (P5 n = 3, P21 n = 2) and Gsdmd (P5 n = 3, P21 n = 2) using RNAscope. c, Quantification of Casp1 (P5 n = 4, P21 n = 2), Aim2 (P5 n = 3, P21 n = 2) and Gsdmd (P5 n = 3, P21 n = 2) mRNA puncta in the hippocampus per 40× image; from one experiment. n values refer to biological replicates. Data are mean ± s.e.m.

Source Data

Extended Data Fig. 2 ASC speck formation routinely occurs in the developing brain but is rare in mature lymph nodes under steady-state conditions.

a, Top, sagittal image of ASC speck formation (green) in the brain of P5 ASC-citrine reporter mice. Original magnification, ×10. Bottom, ASC specks are detected throughout the brain using a ×40 objective including in the cerebellum (i), midbrain (ii), hippocampus (iii) and thalamus (iv). Representative images from three independent experiments with similar results. b, c, Adult (8–12 weeks old) ASC-citrine reporter mice were evaluated for peripheral inflammasome activation based on ASC speck formation (green) in the deep cervical lymph node (DCLN) using confocal microscopy with a ×10 objective (b). c, Arrow shows magnified image of ASC speck (green) formed in the DCLN. Representative images from two independent experiments with similar results.

Extended Data Fig. 3 Lack of AIM2 inflammasome signalling results in an increase in anxiety-related behaviours but not depressive-related behaviours.

Adult (8–12 weeks old) wild-type, Ice−/−, Nlrp3−/− and Aim2−/− mice were assessed for behavioural abnormalities. a, b, Number of urinations (WT n = 26, Ice−/− n = 17, Nlrp3−/− n = 11, Aim2−/− n = 24; from three independent experiments) (a) and number of faecal pellets (WT n = 20, Ice−/− n = 17, Nlrp3−/− n = 11, Aim2−/− n = 21; from three independent experiments) (b) were measured during 10 min of open-field testing. c, d, Depressive behaviours were evaluated in adult male mice using the tail suspension test for escape behaviour (WT n = 19, Ice−/− n = 10, Nlrp3−/− n = 5, Aim2−/− n = 14; from two independent experiments) (c) and sucrose preference test (WT n = 3, Ice−/− n = 3, Aim2−/− n = 4; from one independent experiment) (d). All n values refer to the number of mice used. Data are mean ± s.e.m. P values were determined by one-way ANOVA with Tukey’s post hoc tests.

Source Data

Extended Data Fig. 4 ASC specks form in response to DNA damage in the developing brain.

a, Brains from wild-type P5 mice were evaluated for localization of ASC specks (green) in relation to DAPI+ nuclei (blue) containing DNA damage (γH2AX (red), 53BP1 (grey)) in the cerebellum. i, ii, Magnified images in regions showing ASC specks formed in close proximity to nuclei containing DNA damage. Representative images from four mice with similar results, from one experiment. Original magnifications, ×40. Differences in the size of nuclei probably reflect specific stages in replication, DNA repair, differentiation, or cell death that the individual cells are in, as well as differences seen across CNS cell types. be, Wild-type and Aim2−/− P5 mice received either control treatment or 14 Gy of ionizing radiation (IR) to induce DNA damage. Brains were obtained 6 h later, and immunostained to measure DNA damage induction (γH2AX staining) and inflammasome activation (ASC speck formation) in the cerebellum. b, Representative cerebellar images of γH2AX staining; from two independent experiments with similar results. Original magnification, ×20. c, Quantification of γH2AX staining in the cerebellum (untreated: WT n = 3, Aim2−/− n = 3; IR-treated: WT n = 8, Aim2−/− n = 8; from two independent experiments). d, Representative cerebellar images of ASC speck formation; from two independent experiments with similar results. Original magnification, ×20. e, Quantification of ASC speck formation in the cerebellum (untreated: WT n = 7, Aim2−/− n = 5; IR-treated: WT n = 6, Aim2−/− n = 8; from two independent experiments). All n values refer to the number of mice used. Data are mean ± s.e.m. P values were determined by unpaired two-tailed Student’s t-test.

Source Data

Extended Data Fig. 5 Anxiety phenotypes do not develop in mice that lack IL-1R, IL-18R, MYD88 or caspase-11.

All behavioural testing was conducted on adult (8–12 weeks old) mice. ac, Behaviours for anxiety were evaluated by bouts into (a) and time spent in (b) the centre of the open-field arena, and the total distance travelled (c) (WT n = 11, Il1r−/− n = 19, Il18r−/− n = 22, Myd88−/− n = 12; from three independent experiments). d, Anxiety-related behaviours were assessed in wild-type and Myd88−/− mice using the elevated plus maze. Representative heat maps from four independent experiments with similar results depicting path of travel through open and closed arms of the maze. e, Representative heat maps from four independent experiments with similar results of the path travelled by adult wild-type and Myd88−/− mice in the open-field arena. fh, Quantification of bouts into (f) and time spent in (g) the centre of the open-field arena, and the total distance travelled (h) (WT n = 14, Casp11−/− n = 10; from two independent experiments). All n values refer to the number of mice used. Data are mean ± s.e.m. No statistically significant differences were determined by one-way ANOVA with Tukey’s post hoc tests (ac) or unpaired two-tailed Student’s t-test (fh).

Source Data

Extended Data Fig. 6 Genetic ablation of the AIM2 inflammasome or gasdermin-D in CNS cells limits cell death in response to DNA insults.

Mixed neural cultures were generated from wild-type, Ice−/−, Aim2−/− and Gsdmd−/− P0 mice. a, Mixed neural cell cultures were left untreated to test for baseline differences in cytotoxicity (WT n = 5, Ice−/− n = 5, Aim2−/− n = 5, Gsdmd−/− n = 2). b, Mixed neural cell cultures were primed with LPS for 4 h followed by transfection with poly(dA:dT) (WT n = 4, Ice−/− n = 4, Aim2−/− n = 4, Gsdmd−/− n = 4). Cell death was measured by LDH release after overnight stimulation. Representative data from three independent experiments. All n values refer to biological replicates from one representative experiment. Data are mean ± s.e.m. P values were determined by one-way ANOVA with Tukey’s post hoc tests.

Source Data

Extended Data Fig. 7 AIM2 contributes to CNS cell death during neurodevelopment and in response to ionizing radiation.

a, Representative images from wild-type, Ice−/−, Aim2−/− and Gsdmd−/− P5 mice showing TUNEL+ cells (green) in the cerebellum. Images are representative of two independent experiments with similar results. Original magnifications, ×20. b, Representative images of additional markers of cell death (propidium iodide (PI), grey) in wild-type and Aim2−/− P5 mice. Original magnifications, ×20. c, Quantification of propidium iodide-positive cells in the cerebellum of WT (n = 4) and Aim2−/− (n = 4) P5 mice; from one independent experiment. d, e, Wild-type and Aim2−/− P5 mice received either control treatment or 14 Gy of ionizing radiation to induce DNA damage. Brains were obtained 6 h later and the TUNEL assay was conducted on cerebellar sections to evaluate cell death. d, Representative images showing TUNEL staining in the cerebellum of untreated and irradiated wild-type and Aim2−/− P5 mice; from three independent experiments with similar results. Original magnifications, ×20. e, Quantification of TUNEL+ cells in the cerebellums of untreated and irradiated wild-type (n = 11 untreated, n = 9 IR) and Aim2−/− (n = 9 untreated, n = 8 IR) mice; from three independent experiments. All n values refer to the number of mice used. Data are mean ± s.e.m. P values were determined by unpaired two-tailed Student’s t-test.

Source Data

Extended Data Fig. 8 Lack of AIM2 inflammasome components increases the number of Purkinje neurons that are incorporated into the adult brain.

Representative images of cerebellums from adult (8–12 weeks old) wild-type, Ice−/−, Aim2−/− and Gsdmd−/− mice showing an increase in the number of Purkinje cells (calbindin+ cells) in mice lacking inflammasome components. Original magnifications, ×20. Images are representative of three independent experiments with similar results.

Extended Data Fig. 9 Aim2 is expressed by neurons, astrocytes and microglia in the developing brain.

Brains from wild-type P5 mice (n = 3; from 1 experiment) were evaluated for expression of Aim2 using RNAscope. a, Images showing co-expression of Aim2 (green) and CNS cell-specific genes Rbfox3: NeuN (red), Gfap: GFAP (red), and Aif1: Iba1 (red) in the hippocampus. Original magnifications, ×40. b, Quantification of the percentage of CNS cells in images that are positive for Aim2. n values refer to biological replicates. Data are mean ± s.e.m.

Source Data

Extended Data Fig. 10 Deletion of caspase-1 in CX3CR1-expressing cells does not result in the development of anxiety-related behaviours.

ac, Adult (8–12 weeks old) Casp1fl/fl (n = 10) and Casp1fl/flCx3cr1cre (n = 11) mice were evaluated for anxiety-related behaviours using the time spent in the open arms (a) and distance travelled (b) in the elevated plus maze, along with total bouts into the centre of the open-field arena (c). Data are from two independent experiments. All n values refer to the number of mice used. d, Schematic of the proposed role that DNA damage surveillance by the AIM2 inflammasome has in neurodevelopment. Data are mean ± s.e.m. P values determined by unpaired two-tailed Student’s t-test show no statistically significant differences. Graphical illustrations were made using BioRender (https://biorender.com/).

Source Data

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lammert, C.R., Frost, E.L., Bellinger, C.E. et al. AIM2 inflammasome surveillance of DNA damage shapes neurodevelopment. Nature 580, 647–652 (2020). https://doi.org/10.1038/s41586-020-2174-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2174-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing