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:

Severe reactive astrocytes precipitate pathological hallmarks of Alzheimer’s disease via H2O2 production

Abstract

Although the pathological contributions of reactive astrocytes have been implicated in Alzheimer’s disease (AD), their in vivo functions remain elusive due to the lack of appropriate experimental models and precise molecular mechanisms. Here, we show the importance of astrocytic reactivity on the pathogenesis of AD using GiD, a newly developed animal model of reactive astrocytes, where the reactivity of astrocytes can be manipulated as mild (GiDm) or severe (GiDs). Mechanistically, excessive hydrogen peroxide (H2O2) originated from monoamine oxidase B in severe reactive astrocytes causes glial activation, tauopathy, neuronal death, brain atrophy, cognitive impairment and eventual death, which are significantly prevented by AAD-2004, a potent H2O2 scavenger. These H2O2-induced pathological features of AD in GiDs are consistently recapitulated in a three-dimensional culture AD model, virus-infected APP/PS1 mice and the brains of patients with AD. Our study identifies H2O2 from severe but not mild reactive astrocytes as a key determinant of neurodegeneration in AD.

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: Astrocyte hypertrophy and dose-modulated reactivity in an astrocyte-specific toxin receptor model (GiD).
Fig. 2: H2O2-mediated microglial activation, nitrosative stress, tauopathy and neurodegeneration in GiDs astrocytes.
Fig. 3: H2O2-mediated permanent cognitive deficits, brain atrophy and decreased survival in GiDs mice.
Fig. 4: H2O2-mediated reactive astrocytes, tau phosphorylation and neurodegeneration in a microfluidic human AD brain model.
Fig. 5: DTR-mediated increase of astrocyte reactivity and its effects on neurodegeneration, spike probability and memory ability in APP/PS1 mice.
Fig. 6: Astrocytic hypertrophy, nitrosative stress, brain inflammation and tauopathy in the brain of patients with AD.

Similar content being viewed by others

Data availability

The datasets generated and/or analyzed during the current study are available in the Mendeley data repository (https://data.mendeley.com/) with DOI https://doi.org/10.17632/8mf35ntz9z.1. The datasets generated for the supplementary figures are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Jack, C. R. Jr. et al. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 9, 119–128 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Sofroniew, M. V. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 32, 638–647 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Anderson, M. A., Ao, Y. & Sofroniew, M. V. Heterogeneity of reactive astrocytes. Neurosci. Lett. 565, 23–29 (2014).

    CAS  PubMed  Google Scholar 

  4. Ben Haim, L., Carrillo-de Sauvage, M. A., Ceyzériat, K. & Escartin, C. Elusive roles for reactive astrocytes in neurodegenerative diseases. Front. Cell. Neurosci. 9, 278 (2015).

    PubMed  PubMed Central  Google Scholar 

  5. Kraft, A. W. et al. Attenuating astrocyte activation accelerates plaque pathogenesis in APP/PS1 mice. FASEB J. 27, 187–198 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kamphuis, W. et al. GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer’s disease. Glia 63, 1036–1056 (2015).

    PubMed  Google Scholar 

  7. Sirko, S. et al. Reactive glia in the injured brain acquire stem cell properties in response to Sonic hedgehog. Cell Stem Cell 12, 426–439 (2013).

    CAS  PubMed  Google Scholar 

  8. Chun, H. & Lee, C. J. Reactive astrocytes in Alzheimer’s disease: a double-edged sword.Neurosci. Res. 126, 44–52 (2018).

    CAS  PubMed  Google Scholar 

  9. Chun, H., Marriott, I., Lee, C. J. & Cho, H. Elucidating the interactive roles of glia in Alzheimer’s disease using established and newly developed experimental models. Front. Neurol. 9, 797 (2018).

    PubMed  PubMed Central  Google Scholar 

  10. Ben Haim, L. et al. The JAK/STAT3 pathway is a common inducer of astrocyte reactivity in Alzheimer’s and Huntington’s diseases. J. Neurosci. 35, 2817–2829 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Allaman, I. et al. Amyloid-β aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability. J. Neurosci. 30, 3326–3338 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Hertz, L., Peng, L. & Dienel, G. A. Energy metabolism in astrocytes: high rate of oxidative metabolism and spatiotemporal dependence on glycolysis/glycogenolysis. J. Cereb. Blood Flow. Metab. 27, 219–249 (2007).

    CAS  PubMed  Google Scholar 

  14. Pomilio, C. et al. Glial alterations from early to late stages in a model of Alzheimer’s disease: evidence of autophagy involvement in Aβ internalization. Hippocampus 26, 194–210 (2016).

    CAS  PubMed  Google Scholar 

  15. Ries, M. & Sastre, M. Mechanisms of Aβ clearance and degradation by glial cells. Front. Aging Neurosci. 8, 160 (2016).

    PubMed  PubMed Central  Google Scholar 

  16. Wyss-Coray, T. et al. Adult mouse astrocytes degrade amyloid-β. in vitro and in situ. Nat. Med. 9, 453–457 (2003).

    CAS  PubMed  Google Scholar 

  17. Korenić, A., Andjus, P., Radenović, L. & Spasojević, I. The role of autophagy and lipolysis in survival of astrocytes under nutrient deprivation. Neurosci. Lett. 595, 128–133 (2015).

    PubMed  Google Scholar 

  18. Jo, S. et al. GABA from reactive astrocytes impairs memory in mouse models of Alzheimer’s disease. Nat. Med. 20, 886–896 (2014).

    CAS  PubMed  Google Scholar 

  19. Chen, Q. M., Tu, V. C., Wu, Y. & Bahl, J. J. Hydrogen peroxide dose dependent induction of cell death or hypertrophy in cardiomyocytes. Arch. Biochem. Biophys. 373, 242–248 (2000).

    CAS  PubMed  Google Scholar 

  20. Afanas’ev, I. New nucleophilic mechanisms of ROS-dependent epigenetic modifications: comparison of aging and cancer. Aging Dis. 5, 52–62 (2014).

    PubMed  Google Scholar 

  21. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat. Methods 2, 419–426 (2005).

    CAS  PubMed  Google Scholar 

  22. Park, Y. M., Chun, H., Shin, J. I. & Lee, C. J. Astrocyte specificity and coverage of hGFAP-CreERT2 [Tg(GFAP-Cre/ERT2)13Kdmc] mouse line in various brain regions. Exp. Neurobiol. 27, 508–525 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Gropp, E. et al. Agouti-related peptide-expressing neurons are mandatory for feeding. Nat. Neurosci. 8, 1289–1291 (2005).

    CAS  PubMed  Google Scholar 

  24. Durieux, P. F. et al. D2R striatopallidal neurons inhibit both locomotor and drug reward processes. Nat. Neurosci. 12, 393–395 (2009).

    CAS  PubMed  Google Scholar 

  25. Arruda-Carvalho, M. et al. Posttraining ablation of adult-generated olfactory granule cells degrades odor–reward memories. J. Neurosci. 34, 15793–15803 (2014).

    PubMed  PubMed Central  Google Scholar 

  26. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ghosh, A. et al. Targeted ablation of oligodendrocytes triggers axonal damage. PLoS ONE 6, e22735 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Chun, H. et al. Astrocytic proBDNF and Tonic GABA distinguish active versus reactive astrocytes in hippocampus. Exp. Neurobiol. 27, 155–170 (2018).

    PubMed  PubMed Central  Google Scholar 

  29. Maggiorani, D. et al. Monoamine oxidases, oxidative stress, and altered mitochondrial dynamics in cardiac ageing. Oxid. Med. Cell. Longev. 2017, 3017947 (2017).

    PubMed  PubMed Central  Google Scholar 

  30. Shin, J. H. et al. Concurrent blockade of free radical and microsomal prostaglandin E synthase-1-mediated PGE2 production improves safety and efficacy in a mouse model of amyotrophic lateral sclerosis. J. Neurochem. 122, 952–961 (2012).

    CAS  PubMed  Google Scholar 

  31. Borroni, E. et al. Sembragiline: a novel, selective monoamine oxidase type B inhibitor for the treatment of Alzheimer’s disease. J. Pharmacol. Exp. Ther. 362, 413–423 (2017).

    CAS  PubMed  Google Scholar 

  32. Zhu, D. et al. Hydrogen peroxide alters membrane and cytoskeleton properties and increases intercellular connections in astrocytes. J. Cell Sci. 118, 3695–3703 (2005).

    CAS  PubMed  Google Scholar 

  33. Park, J.-H. et al. Newly developed reversible MAO-B inhibitor circumvents the shortcomings of irreversible inhibitors in Alzheimer’s disease.Sci. Adv. 5, eaav0316 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Popa-Wagner, A., Mitran, S., Sivanesan, S., Chang, E. & Buga, A.-M. ROS and brain diseases: the good, the bad, and the ugly. Oxid. Med. Cell. Longev. 2013, 963520 (2013).

    PubMed  PubMed Central  Google Scholar 

  35. Garwood, C. J., Pooler, A. M., Atherton, J., Hanger, D. P. & Noble, W. Astrocytes are important mediators of Aβ-induced neurotoxicity and tau phosphorylation in primary culture. Cell Death Dis. 2, e167 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Krstic, D. et al. Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J. Neuroinflammation 9, 151 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Eidet, J. R., Pasovic, L., Maria, R., Jackson, C. J. & Utheim, T. P. Objective assessment of changes in nuclear morphology and cell distribution following induction of apoptosis. Diagn. Pathol. 9, 92 (2014).

    PubMed  PubMed Central  Google Scholar 

  38. Benzing, W. C., Mufson, E. J. & Armstrong, D. M. Alzheimer’s disease-like dystrophic neurites characteristically associated with senile plaques are not found within other neurodegenerative diseases unless amyloid β-protein deposition is present. Brain Res. 606, 10–18 (1993).

    CAS  PubMed  Google Scholar 

  39. Yang, D.-S. et al. Neuronal apoptosis and autophagy cross talk in aging PS/APP mice, a model of Alzheimer’s disease. Am. J. Pathol. 173, 665–681 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Augustinack, J. C., Schneider, A., Mandelkow, E.-M. & Hyman, B. T. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathol. 103, 26–35 (2002).

    CAS  PubMed  Google Scholar 

  41. Stephenson, R. M. & Andrew, R. J. Amnesia due to β-antagonists in a passive avoidance task in the chick. Pharmacol. Biochem. Behav. 15, 597–604 (1981).

    CAS  PubMed  Google Scholar 

  42. Park, J. et al.A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease.Nat. Neurosci. 21, 941–951 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Schwab, C., Klegeris, A. & McGeer, P. L. Inflammation in transgenic mouse models of neurodegenerative disorders. Biochim. Biophys. Acta 1802, 889–902 (2010).

    CAS  PubMed  Google Scholar 

  44. Ortinski, P. I. et al. Selective induction of astrocytic gliosis generates deficits in neuronal inhibition. Nat. Neurosci. 13, 584–591 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Haettig, J., Sun, Y., Wood, M. A. & Xu, X. Cell-type specific inactivation of hippocampal CA1 disrupts location-dependent object recognition in the mouse. Learn. Mem. 20, 139–146 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Woo, J. et al. Functional characterization of resting and adenovirus-induced reactive astrocytes in three-dimensional culture. Exp. Neurobiol. 26, 158–167 (2017).

    PubMed  PubMed Central  Google Scholar 

  47. Liu, P. P., Xie, Y., Meng, X.-Y. & Kang, J.-S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target. Ther. 4, 29 (2019).

    PubMed  PubMed Central  Google Scholar 

  48. Hawkes, N. Merck ends trial of potential Alzheimer’s drug verubecestat. BMJ 356, j845 (2017).

    PubMed  Google Scholar 

  49. Roostaei, T. et al. Genome-wide interaction study of brain beta-amyloid burden and cognitive impairment in Alzheimer’s disease. Mol. Psychiatry 22, 287–295 (2017).

    CAS  PubMed  Google Scholar 

  50. Lee, C. J. et al. Astrocytic control of synaptic NMDA receptors. J. Physiol. 581, 1057–1081 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim, Y. et al. Uvrag targeting by Mir125a and Mir351 modulates autophagy associated with Ewsr1 deficiency. Autophagy 11, 796–811 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Choi, S. H. et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515, 274–278 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by the Creative Research Initiative Program, Korean National Research Foundation (NRF) (grant no. 2015R1A3A2066619), Brain Research Program through the NRF funded by the Ministry of Science and ICT (grant nos. 2018M3C7A1056682 and 2018M3C7A1056897), a National Research Council of Science & Technology grant by the Korean government (MSIP) (no. CRC-15-04-KIST), grant no. 2E28411 from the Korean Institute of Science and Technology (KIST) and grant no. IBS-R001-D2 from the Institute for Basic Science from the Ministry of Science (to C.J.L.). This study was also supported by a National Institutes of Health grant no. AG054156, grant no. NRF-2016M3C7A1904233 and grant no. 2E26663 from KIST (to H.R.). This work was supported by the Pioneering Funding Award funded by the Cure Alzheimer’s Fund and the NIH (grant no. AG059236-01A1 to H.C.), by the National Honor Scientist Program (grant no. NRF-2012R1A3A1050385 to B.K.K.) and by The Bio & Medical Technology Development Program (grant no. NRF‐2019M3E5D2A01066259 to D.K.).

Author information

Authors and Affiliations

Authors

Contributions

H.J.C., H.R. and C.J.L. designed the study, analyzed the data and wrote the manuscript. H.J.C., J.W.L. and Y.M.P. performed and analyzed the IHC in GiD, fGiD and APP/PS1 mice. Y.H.K. performed and analyzed the western blot and MTT assay. Y.J.K. performed and analyzed the experiments in the 3D neurocyte AD model. H.J.C. and J.W.L. performed the microdialysis and in vivo H2O2 assay. W.J.W. performed the IHC and in vitro H2O2 assay. Y.M.P. validated the GFAP-CreERT2 mice. J.W.L., J.H.S., Y.H.J. and S.K. performed the behavioral tests. H.J.C., W.J.W., Y.H.J., J.K.L. and J.S.W. performed the electrophysiological experiments. S.E.L. generated the adenovirus and AAV virus. H.J.I. and Y.J.H. performed the immunostaining and quantitative RT–PCR analysis in the human samples. D.Y.K. and H.C. provided the 3D neurocyte AD model. J.H.S. and B.J.G. provided the AAD-2004 scavenger. S.M.J. and D.K. provided the transgenic animal models. Y.S.K. provided the Aβ. J.H.P. and K.D.P. provided the KDS2010 MAO-B inhibitor. H.S.C. and J.S.S. performed the gene expression analysis. B.K.K. designed the experiments and wrote the manuscript. All authors contributed to the analysis and discussion of the results and reviewed the manuscript.

Corresponding authors

Correspondence to Hoon Ryu or C. Justin Lee.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Validation and characterization of GiD system.

a, (left) RT-PCR analysis of diphtheria toxin receptor(DTR) in hippocampus of Gcon and GiDm. (right) RT-PCR analysis of DTR in cultured astrocytes from Gcon and GiD (GiDc). GAPDH, internal control. Experiments were repeated more than twice. b, Measuring passive conductance in astrocytes of Gcon and GiDs. c, Experimental procedures of GiD mice, which GFAP-CreERT2 crossed with iDTR mice. Tam, tamoxifen, 100 mg/kg/day, 5 days; DT, 50 µg/kg/day, 2 days. d, GFAP immunostaining in brain regions of frontal lobe containing cortex, hippocampus, striatum and amygdala of Gcon and GiD mice. Ctx, cortex; Hipp, hippocampus; Str, striatum; Amyg, amygdala. e, Experimental procedures for PI staining in GiDm, CiD mice and KA-injected seizure model mice. DT, 50 µg/kg/day, 2 days; KA, kainic acid, 25 mg/kg. f, PI staining in GiDm, CiD and seizure model mice. g, h, MTT assay in DT-treated GiD astrocyte. DT, 1 ug/ml, 5 days; 3-MA, 3-Methyladenine, 0.5 mM (g), Baf A1, bafilomycin A1, 4 µM (h). Data are presented as mean ± SEM. *P < 0.05; NS: not significant. Additional statistical details are provided in Supplementary Table 1.

Source data

Extended Data Fig. 2 Autophagy activation in astrocytes of GiDs and GiDc.

a, Schematic diagram for possible mechanism of DT intoxification in GiD. b, Immunostaining for LC3 and GFAP in cortex of Gcon and GiDs. c, d, Immunostaining for LC3 (c) and SQSTM1 (d) in hippocampus of Gcon and GiDs. e, Experimental timeline for DT treatment in cultured astrocytes from Gcon and GiD (GiDc). f, g, Western blot analysis showing that levels of endogenous SQSTM1 and LC3-II are time-dependently increased in DT-treated GiDc astrocyte. h-s, Autophagic flux assay and densitometry analysis in DT-treated GiDc astrocytes. DT, 1 µg/ml, 8 hrs; E64d+Pep.A, 10 µg/ml E64d plus 10 µg/ml Pep.A, 6 hrs (h-m); CQ, 20 µM, 6 hrs (n-s). Data are presented as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; NS: not significant. Additional statistical details are provided in Supplementary Table 1.

Source data

Extended Data Fig. 3 MAO-B-mediated astrocytic hypertrophy and lack of neurodegeneration in GiDm brain.

a, Experimental procedures for AAD-2004 and selegiline treatment in GiDm. b,c, Immunostaining and quantification for GFAP in Gcon, GiDm, selegline-treated GiDm and AAD-2004-treated GiDm. d,e, Immunostaining and quantification for NeuN in Gcon and GiDm. Data are presented as mean ± SEM. ***P < 0.001, ****P < 0.0001; NS: not significant. Additional statistical details are provided in Supplementary Table 1.

Extended Data Fig. 4 Astrocytic hypertrophy and neurodegeneration in cortex, striatum and amygdala of GiDs brain.

a, Representative image of immunostaining for GFAP and NeuN in the brain of Gcon and GiDs. b-d, Quantification for the number of cells positive for NeuN in cortex, striatum and amygdala of Gcon and GiDs. Data are presented as mean ± SEM. *P < 0.05, ***P < 0.001, ****P < 0.0001. Additional statistical details are provided in Supplementary Table 1.

Extended Data Fig. 5 Development of focal GiD (fGiD).

a, b, Schematic diagram and experimental procedure for making mild and severe focal GiD, fGiDm and fGiDs, respectively. c, Immunostaining for GFAP and mCherry in CTL, fGiDm and fGiDs. d, Quantification for GFAP intensity in CTL, fGiDm and fGiDs. e, DAPI image in CTL, fGiDm and fGiDs. f, Counting the number of DAPI signals in stratum radiatum of CTL, fGiDm and fGiDs. g, Measuring the size of CA1 region in CTL, fGiDm and fGiDs. h, Comparison of glial activation in hippocampus between fGiDs and GiDs. Data are presented as mean ± SEM. **P < 0.01, ***P < 0.001, ****P < 0.0001; NS: not significant. Additional statistical details are provided in Supplementary Table 1.

Extended Data Fig. 6 Virus-mediated increase of astrocyte’s reactivity and its effects on neurodegeneration and tauopathy in APP/PS1 mice.

a, Schematic diagram and experimental timeline for increasing the reactivity of astrocytes in APP/PS1 mice using Adeno-GFAP-GFP virus. b-d, Immunostaining for GFAP, NeuN and quantification of the mean intensity in uninjected- or virus injected- WT and APP/PS1 mice. Magenta, Adeno-GFAP-GFP virus. e-g, DAB staining and quantification for p-Tau(S199) and p-Tau(S396) in virus injected- WT and APP/PS1 mice. Data are presented as mean ± SEM. *P < 0.05, **P < 0.01, ****P < 0.0001; NS: not significant. Additional statistical details are provided in Supplementary Table 1.

Supplementary information

Supplementary Information

Supplementary Figs. 1–11 and Supplementary Tables 1 and 2.

Reporting Summary

Source data

Source Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 2

Unprocessed western blots and/or gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chun, H., Im, H., Kang, Y.J. et al. Severe reactive astrocytes precipitate pathological hallmarks of Alzheimer’s disease via H2O2 production. Nat Neurosci 23, 1555–1566 (2020). https://doi.org/10.1038/s41593-020-00735-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-020-00735-y

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