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.

  • Article
  • Published:

Activation of innate immune cGAS-STING pathway contributes to Alzheimer’s pathogenesis in 5×FAD mice

Abstract

cGAS senses microbial and host-derived double-stranded DNA in cytoplasm to trigger cellular innate immune response in a STING-dependent manner; however, it remains unknown whether the cGAS-STING pathway in innate immunity contributes to Alzheimer’s disease (AD). Here we demonstrated the detectable binding of the cGAS double-stranded DNA in cytoplasm and the activation of the microglial cGAS-STING pathway in brains of human AD and aged mice. Cgas−/−;5×FAD mice were largely protected from cognitive impairment, amyloid-β pathology, neuroinflammation and other sequelae associated with AD. Furthermore, Cgas deficiency in microglia inhibited a neurotoxic A1 astrocytic phenotype and thus alleviated oligomeric amyloid-β peptide-induced neurotoxicity. Finally, administration of STING inhibitor H-151 potently suppressed the activation of the cGAS-STING pathway and ameliorated AD pathogenesis in 5×FAD mice. In conclusion, our present study has identified a critical molecular link between innate immunity and AD and suggests that therapeutic targeting of the cGAS-STING pathway activity might effectively interfere with the progression of AD.

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

Access options

Buy this article

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

Fig. 1: cGAS-STING pathway is activated in brains during AD and aging.
Fig. 2: cGAS deficiency alleviates cognitive decline, Aβ pathology and neuroinflammation in 5×FAD mice.
Fig. 3: The suppression of neurotoxic A1 astrocytes by cGAS deficient microglia is neuroprotective from Aβ toxicity.
Fig. 4: Administration of STING inhibitor H-151 ameliorates Aβ pathology and neuroinflammation in 5×FAD mice.

Similar content being viewed by others

Data availability

All data necessary to understand and evaluate the conclusion of this paper are provided in the article, its Source Data and Supplementary Information. Any other data can be obtained from the corresponding author upon reasonable request.

References

  1. Ablasser, A. & Chen, Z. J. cGAS in action: expanding roles in immunity and inflammation. Science 363, eaat8657 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Ablasser, A. et al. cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498, 380–384 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gao, P. et al. Cyclic [G(2′,5′) pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell 153, 1094–1107 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ishikawa, H., Ma, Z. & Barber, G. N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 461, 788–792 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sun, L. J., Wu, J. X., Du, F. H., Chen, X. & Chen, Z. J. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Wu, J. X. et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Cai, X., Chiu, Y. H. & Chen, Z. J. The cGAS-cGAMP-STING pathway of cytosolic DNA sensing and signaling. Mol. Cell 54, 289–296 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Civril, F. et al. Structural mechanism of cytosolic DNA sensing by cGAS. Nature 498, 332–337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Li, T. & Chen, Z. J. J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 215, 1287–1299 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tan, X. J., Sun, L. J., Chen, J. Q. & Chen, Z. J. J. Detection of microbial infections through innate immune sensing of nucleic acids. Annu. Rev. Microbiol. 72, 447–478 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Chen, Q., Sun, L. J. & Chen, Z. J. J. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat. Immunol. 17, 1142–1149 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Marcus, A. et al. Tumor-derived cGAMP triggers a STING-mediated interferon response in non-tumor cells to activate the NK cell response. Immunity 49, 754–763 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dou, Z. X. et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550, 402–406 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gluck, S. et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol. 19, 1061–1070 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yang, H., Wang, H. Z., Ren, J. Y., Chen, Q. & Chen, Z. J. J. cGAS is essential for cellular senescence. Proc. Natl Acad. Sci. USA 114, E4612–E4620 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gui, X. et al. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature 567, 262–266 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gao, D. X. et al. Activation of cyclic GMP-AMP synthase by self-DNA causes autoimmune diseases. Proc. Natl Acad. Sci. USA 112, E5699–E5705 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, Q. et al. Inhibition of double-strand DNA-sensing cGAS ameliorates brain injury after ischemic stroke. EMBO Mol. Med. 12, e11002 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sliter, D. A. et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258–262 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sharma, M., Rajendrarao, S., Shahani, N., Ramirez-Jarquin, U. N. & Subramaniam, S. Cyclic GMP-AMP synthase promotes the inflammatory and autophagy responses in Huntington disease. Proc. Natl Acad. Sci. USA 117, 15989–15999 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Yu, C. H. et al. TDP-43 triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS. Cell 183, 636–649 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Decout, A., Katz, J. D., Venkatraman, S. & Ablasser, A. The cGAS-STING pathway as a therapeutic target in inflammatory diseases. Nat. Rev. Immunol. 21, 548–569 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hou, Y. J. et al. NAD plus supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc. Natl Acad. Sci. USA 118, e2011226118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Jin, M. et al. Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nat. Commun. 12, 6565 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Querfurth, H. W. & LaFerla, F. M. Alzheimer’s disease. N. Engl. J. Med. 362, 329–344 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Suberbielle, E. et al. Physiologic brain activity causes DNA double-strand breaks in neurons, with exacerbation by amyloid-β. Nat. Neurosci. 16, 613–621 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bozner, P. et al. The amyloid β protein induces oxidative damage of mitochondrial DNA. J. Neuropathol. Exp. Neurol. 56, 1356–1362 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Moya, G. E., Rivera, P. D. & Dittenhafer-Reed, K. E. Evidence for the role of mitochondrial DNA release in the inflammatory response in neurological disorders. Int. J. Mol. Sci. 22, 7030 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Reinert, L. S. et al. Brain immune cells undergo cGAS/STING-dependent apoptosis during herpes simplex virus type 1 infection to limit type IIFN production. J. Clin. Invest. 131, e136824 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Reinert, L. S. et al. Sensing of HSV-1 by the cGAS-STING pathway in microglia orchestrates antiviral defence in the CNS. Nat. Commun. 7, 13348 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pascoal, T. A. et al. Microglial activation and tau propagate jointly across Braak stages. Nat. Med. 27, 2048–2049 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Oakley, H. et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 26, 10129–10140 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu, H. P. et al. Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature 563, 131–136 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. d’Errico, P. et al. Microglia contribute to the propagation of Aβ into unaffected brain tissue. Nat. Neurosci. 25, 20–25 (2022).

    Article  PubMed  Google Scholar 

  35. Shi, Y. et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549, 523–527 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Liang, X. Y. et al. Microglia and its genetics in Alzheimer’s disease. Curr. Alzheimer Res. 18, 676–688 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yun, S. P. et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nat. Med. 24, 931–938 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Griciuc, A. et al. TREM2 acts downstream of CD33 in modulating microglial pathology in Alzheimer’s disease. Neuron 103, 820–835 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hou, Y. et al. NAD(+) supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc. Natl Acad. Sci. USA 118, e2011226118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Leng, F. & Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat. Rev. Neurol. 17, 157–172 (2021).

    Article  PubMed  Google Scholar 

  42. Roy, E. R. et al. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. J. Clin. Invest. 130, 1912–1930 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pascoal, T. A. et al. Microglial activation and tau propagate jointly across Braak stages. Nat. Med. 27, 1592–1599 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. Heneka, M. T. et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493, 674–678 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Ising, C. et al. NLRP3 inflammasome activation drives tau pathology. Nature 575, 669–673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Luteijn, R. D. et al. SLC19A1 transports immunoreactive cyclic dinucleotides. Nature 573, 434–438 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhou, C. et al. Transfer of cGAMP into bystander cells via LRRC8 volume-regulated anion channels augments STING-mediated interferon responses and anti-viral immunity. Immunity 52, 767–781 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Ablasser, A. et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sil, A. et al. Sex differences in behavior and molecular pathology in the 5×FAD model. J. Alzheimers Dis. 85, 755–778 (2022).

    Article  PubMed  Google Scholar 

  50. Sagare, A. P. et al. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nat. Commun. 4, 2932 (2013).

    Article  PubMed  Google Scholar 

  51. Deacon, R. M. J. Assessing nest building in mice. Nat. Protoc. 1, 1117–1119 (2006).

    Article  PubMed  Google Scholar 

  52. Zeng, J. X. et al. TRIM9-mediated resolution of neuroinflammation confers neuroprotection upon ischemic stroke in mice. Cell Rep. 27, 549–560 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zeng, J. X. et al. The zika virus capsid disrupts corticogenesis by suppressing dicer activity and miRNA biogenesis. Cell Stem Cell 27, 618–632 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zeng, J. et al. Specific inhibition of the NLRP3 inflammasome suppresses immune overactivation and alleviates COVID-19 like pathology in mice. eBioMedicine 75, 103803 (2022).

    Article  CAS  PubMed  Google Scholar 

  55. Kim, J. et al. VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease. Science 366, 1531–1536 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank B. Lu from the Central South University in China for providing additional Cgas−/− mice and Y.-H. He from the Kunming Institute of Zoology, Chinese Academy of Sciences for providing partial aged mice samples. The graphical diagram is produced by BioRender. This work was supported by Yunnan Fundamental Research Projects (grant no. 202201AW070020).

Author information

Authors and Affiliations

Authors

Contributions

All authors read and approved the final version of the manuscript. J.Z. and Z.Z. conceived the research and designed the study. J.Z. and Z.Z. wrote the manuscript. X.X., G.M., X.L. and J.B.Z. performed the experiments and discussed the data. All authors commented on the manuscript.

Corresponding authors

Correspondence to Zhen Zhao or Jianxiong Zeng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Marco Colonna, Hitoshi Okazawa, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Yahyah Aman, in collaboration with the Nature Aging team.

Additional information

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

Extended data

Extended Data Fig. 1 The binding partner of cGAS and cytosolic or nucleus dsDNA in the brain of human AD and aged mice.

a, Quantification of cmtDNA by qPCR in brain cells of 2-month-old 5 × FAD mice and age-matched WT mice. b, Quantification of total cellular mtDNA by qPCR in brain cells of 6-month-old 5 × FAD mice and age-matched WT mice. D-loop indicated a specific fragment within mtDNA. Mean ± SD; n = 4 or 6; n.s., not significant, two-tailed Student’s t-test. c, PLA as indicated by white arrow showing the association of cGAS protein with cytosolic or nucleus DNA in hippocampal tissues of human AD samples. Age-matched health human samples were used as control. Scale bar, 10 μm. d, Quantification of PLA red dots per cell in c. Three sections in each animal were used to count the dots. Mean ± SD; n = 3, three sections of each brain sample were stained and counted; ***, p < 0.001, two-tailed Student’s t-test. e, f, Quantification of cmtDNA (e) and mtDNA (f) by qPCR in brain cells of 20-month-old WT mice. 3-month-old WT mice were used as control. Mean ± SD; n.s., not significant; n = 3; *, p < 0.05, **, p < 0.01, ***, p < 0.001, two-tailed Student’s t-test. g, PLA showing the association of cGAS protein with cytosolic or nucleus DNA in hippocampal tissues of 20-month-old aged WT mice. 3-month-old young WT mice were used as control. Scale bar, 10 μm. h, Quantification of PLA red dots per cell in g. Mean ± SD; n = 3, four sections of each brain sample were stained and counted; ***, p < 0.001, two-tailed Student’s t-test. i, Western immunoblotting analysis of the expression of the indicated proteins involved in cGAS-SITNG pathway of cortical tissues in 20-month-old aged WT mice. n = 3. j, Quantification of the expression of the phosphorylated STING (p-STING), TBK1 (p-TBK1), p65 (p-p65), and IRF3 (p-IRF3) relative to β-actin in i. Mean ± SD; n = 3; *, p < 0.05, **, p < 0.01, ***, p < 0.001, two-tailed Student’s t-test.

Source data

Extended Data Fig. 2 cGAS deficiency ameliorates cognitive decline and reduces microglial activation in 5 × FAD mice.

a, b, Immunostaining (a) and quantification (b) of Aβ in DG of 2~2.5 months old Cgas+/+;5 × FAD and Cgas−/−;5 × FAD mice. Scale bar, 40 μm. Mean ± SD; n = 3; n.s., not significant, two-tailed Student’s t-test. c, Behavioral analysis of burrowing in 6-month-old Cgas+/+;5 × FAD and Cgas–/–;5 × FAD mice. Age-matched Cgas+/+ and Cgas–/– mice were used as control. Mean ± SD; n = 12; ***, p < 0.001, one-way ANOVA with Bonferroni’s post hoc test. d, e, Quantification of Aβ plaque numbers in different plaque diameter (d) and percentage of Aβ plaque area (e) in DG in Fig. 2h. Mean ± SD; n = 6; *, p < 0.05, **, p < 0.01, two-tailed Student’s t-test. f, Quantification of percentage of Iba1+ area in Fig. 2h. Mean ± SD; n = 6; **, p < 0.01, two-tailed Student’s t-test. g, Western immunoblotting analysis of the expression of synaptic PSD95 and Synaptophysin of cortical tissues in 6-month-old WT, Cgas+/+;5 × FAD, and Cgas–/–;5 × FAD mice. n = 3. h, Quantification of protein levels of PSD95 and Synaptophysin relative to β-actin in g. Mean ± SD; n = 3; *, p < 0.05, ***, p < 0.001, two-tailed Student’s t-test.

Source data

Extended Data Fig. 3 cGAS-STING pathway is differentially activated in multiple types of neural cells.

a-f, Quantification of cmtDNA (a-c) and total mtDNA (d-f) in primary cultured microglia (a, d), neurons (b, e), and astrocytes (c, f) treated with oligomeric Aβ42 (5 μM) for 24 h. g-i, ELISA analysis of 2′3′-cGAMP level in oligomeric Aβ42-treated primary microglia (g), neurons (h), and astrocytes (i). j-l, ELISA analysis of IFN-β level in the supernatants of primary microglia (j), neurons (k), and astrocytes (l) treated with oligomeric Aβ42. Mean ± SD; n = 3; n.s., not significant, *, p < 0.05, **, p < 0.01, two-tailed Student’s t-test.

Source data

Extended Data Fig. 4 Additional control experiments of primary neuronal cultures.

ac, Cgas+/+ and Cgas–/– primary neurons that were pre-infected with AAV expressing a GFP with CaMKII promotor were treated with oligomeric Aβ42 (750 nM) for 24 h (a). Fluorescent signals (b) and percentage (c) of GFP-labeled primary neurons were recorded and calculated. Scale bar, 50 μm. d-f, Cgas+/+ primary neurons that were incubated with Cgas+/+ ACM and Cgas–/– ACM without the prior MCM treatment were treated with oligomeric Aβ42 (750 nM) for 24 h (d). Fluorescent signals (e) and percentage (f) of GFP-labeled primary neurons were recorded and calculated. Scale bar, 50 μm. g-i, Cgas+/+ primary neurons that were incubated with Cgas+/+ MCM and Cgas–/– MCM without the ACM treatment were treated with oligomeric Aβ42 (750 nM) for 24 h (g). Scale bar, 50 μm. Fluorescent signals (h) and percentage (i) of GFP-labeled primary neurons were recorded and calculated. In c, f and i, Mean ± SD, n = 4, n.s., not significant by one-way ANOVA with Bonferroni’s post hoc test.

Source data

Extended Data Fig. 5 The purification of astrocytes from brains of mice.

a, Astrocytic cells were magnetically purified by ACSA-2 MicroBeads from brains of mice. b, c, Gating strategy (b) and purity (c) of ACSA-2+ astrocyte populations by anti-ACSA-2-PE antibody in fluorescence activated cell sorter (FACS) analysis. d, Immunofluorescence analysis of purified astrocytes in cultures using anti-GFAP antibody. Scale bar, 50 μm. Representative images as shown were from the results of five mice each group. e, Transcriptional analysis for astrocytic genes in 6-month-old Cgas+/+ (n = 5) and Cgas–/– (n = 5) mice. f, GFAP+ astrocytic staining of DG in 6-month-old Cgas+/+;5 × FAD and Cgas–/–;5 × FAD mice. Scale bar, 50 μm. g, Quantification of percentage of GFAP+ area in f. Mean ± SD; n = 6; **, p < 0.01, two-tailed Student’s t-test.

Source data

Extended Data Fig. 6 The effect of cGAS-STING inhibitors-treated microglia on primary neuronal survival.

a-c, Cgas+/+ primary neurons that were incubated with RU.521 MCM and H-151 MCM without the ACM treatment were treated with oligomeric Aβ42 (750 nM) for 24 h (a). Fluorescent signals (b) and percentage (c) of GFP-labeled primary neurons were recorded and calculated. Mean ± SD, n = 4, n.s., not significant by one-way ANOVA with Bonferroni’s post hoc test.

Source data

Extended Data Fig. 7 H-151 treatment suppressed the activation of cGAS-STING pathway and the expression of the inflammatory genes.

a, Western immunoblotting analysis of the expression of the indicated proteins involved in cGAS-SITNG pathway of cortical tissues in H-151-treated 5month-old 5 × FAD mice. n = 3. b, Quantification of the expression of the phosphorylated TBK1 (p-TBK1), phosphorylated p65 (p-p65) and phosphorylated IRF3 (p-IRF3) relative to β-actin in a. Mean ± SD; n = 3; **, p < 0.01, ***, p < 0.001, two-tailed Student’s t-test. c-f, qRT-PCR analysis of a panel of neuroinflammatory genes in cortical tissues of H-151-treated 5 × FAD mice at 5 months old. Mean ± SD; n = 4; *, p < 0.05, **, p < 0.01, two-tailed Student’s t-test.

Source data

Extended Data Fig. 8 Additional phenotypes of the H-151 treated 5 × FAD mice.

a, Immunostaining of NeuN and Aβ of DG regions in H-151-treated 5-month-old 5 × FAD mice. Scale bar, 50 μm. b, c, Quantification of percentage of NeuN+ area (b) and NeuN+ thickness (c) as indicated by white arrow in a. Mean ± SD; n = 6; n.s., not significant, Student’s t-test. d, Immunostaining of Aβ and CD68 of DG regions in H-151-treated 5-month-old 5 × FAD mice. Scale bar, 50 μm. e, Quantification of CD68/Aβ ratio in d. Mean ± SD; n = 6; **, p < 0.01, two-tailed Student’s t-test.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Information on frozen or paraffin-embedded blocks of postmortem human frontal lobe or hippocampus samples, respectively. Supplementary Table 2 Primers used for analysis in this study. Supplementary Table 3 Antibodies used in this study.

Source data

Source Data

Uncropped blots for Figs. 1d, 2j and 3b and Extended Data Figs.1i, 2g and 7a.

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xie, X., Ma, G., Li, X. et al. Activation of innate immune cGAS-STING pathway contributes to Alzheimer’s pathogenesis in 5×FAD mice. Nat Aging 3, 202–212 (2023). https://doi.org/10.1038/s43587-022-00337-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-022-00337-2

This article is cited by

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