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:

Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration

23 February 2024 Editor's Note: Readers are alerted that concerns have been raised about the data and statistical analysis reported in this article. Further editorial action will be taken if appropriate once the investigation into the concerns is complete and all parties have been given an opportunity to respond in full.

An Author Correction to this article was published on 15 December 2020

This article has been updated

Abstract

In neurodegenerative diseases, debris of dead neurons are thought to trigger glia-mediated neuroinflammation, thus increasing neuronal death. Here we show that the expression of neurotoxic proteins associated with these diseases in microglia alone is sufficient to directly trigger death of naive neurons and to propagate neuronal death through activation of naive astrocytes to the A1 state. Injury propagation is mediated, in great part, by the release of fragmented and dysfunctional microglial mitochondria into the neuronal milieu. The amount of damaged mitochondria released from microglia relative to functional mitochondria and the consequent neuronal injury are determined by Fis1-mediated mitochondrial fragmentation within the glial cells. The propagation of the inflammatory response and neuronal cell death by extracellular dysfunctional mitochondria suggests a potential new intervention for neurodegeneration—one that inhibits mitochondrial fragmentation in microglia, thus inhibiting the release of dysfunctional mitochondria into the extracellular milieu of the brain, without affecting the release of healthy neuroprotective mitochondria.

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: Inhibition of Drp1–Fis1-mediated mitochondrial fission in vivo reduces sustained microglia and astrocyte activation and subsequent proinflammatory responses in three models of neurodegenerative disease in mice.
Fig. 2: Drp1–Fis1-mediated mitochondrial fragmentation and dysfunction are required to induce microglial inflammatory responses in a model of HD (induced by cytotoxic Q73) or LPS activation in culture.
Fig. 3: Drp1–Fis1-mediated mitochondrial fragmentation and dysfunction are required to induce microglial inflammatory response in a model of ALS (by expressing cytotoxic SOD1-G93A) or in a model of AD (induced by treatment with oAβ1-42) in culture.
Fig. 4: Conditioned media of activated microglia with mitochondrial dysfunction propagate astrocyte activation to the A1 proinflammatory state and dysfunction of mitochondria in cultured astrocytes in multiple models of neurodegenerative diseases.
Fig. 5: Excessive mitochondrial fission and dysfunction occur in activated astrocytes.
Fig. 6: Dysfunctional extracellular mitochondria release from activated primary rat and mouse microglia or astrocytes or human primary monocyte-derived microglial cells occurs in a Drp1–Fis1-specific manner.
Fig. 7: Released (extracellular) glial mitochondria propagate mitochondrial dysfunction and cell death from activated microglia to neurons through astrocytes.
Fig. 8: WT or R6/2 astrocytes induce Drp1–Fis1-dependent propagation of neuronal mitochondrial dysfunction and neuronal cell death from R6/2 microglia to naive mouse cortical neurons.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Change history

  • 15 December 2020

    A Correction to this paper has been published: https://doi.org/10.1038/s41593-020-00774-5

  • 23 February 2024

    Editor's Note: Readers are alerted that concerns have been raised about the data and statistical analysis reported in this article. Further editorial action will be taken if appropriate once the investigation into the concerns is complete and all parties have been given an opportunity to respond in full.

References

  1. Aguzzi, A. & O’Connor, T. Protein aggregation diseases: pathogenicity and therapeutic perspectives. Nat. Rev. Drug Discov.9, 237–248 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Hayakawa, K. et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature535, 551–555 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  3. Belanger, M. & Magistretti, P. J. The role of astroglia in neuroprotection. Dialogues Clin. Neurosci.11, 281–295 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Chen, Z. & Trapp, B. D. Microglia and neuroprotection. J. Neurochem.136, 10–17 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Crotti, A. & Glass, C. K. The choreography of neuroinflammation in Huntington’s disease. Trends Immunol.36, 364–373 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Heneka, M. T. et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol.14, 388–405 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Liu, J. & Wang, F. Role of neuroinflammation in amyotrophic lateral sclerosis: cellular mechanisms and therapeutic implications. Front. Immunol.8, 1005 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Ransohoff, R. M. How neuroinflammation contributes to neurodegeneration. Science353, 777–783 (2016).

    Article  CAS  PubMed  ADS  Google Scholar 

  9. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature541, 481–487 (2017).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  10. Itoh, K., Nakamura, K., Iijima, M. & Sesaki, H. Mitochondrial dynamics in neurodegeneration. Trends Cell Biol.23, 64–71 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Reddy, P. H. Increased mitochondrial fission and neuronal dysfunction in Huntington’s disease: implications for molecular inhibitors of excessive mitochondrial fission. Drug Discov. Today19, 951–955 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Joshi, A. U. et al. Inhibition of Drp1/Fis1 interaction slows progression of amyotrophic lateral sclerosis. EMBO Mol. Med.10, e8166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Khalil, B. & Lievens, J. C. Mitochondrial quality control in amyotrophic lateral sclerosis: towards a common pathway? Neural Regen. Res.12, 1052–1061 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Reddy, P. H. et al. Dynamin-related protein 1 and mitochondrial fragmentation in neurodegenerative diseases. Brain Res. Rev.67, 103–118 (2011).

    Article  CAS  PubMed  ADS  Google Scholar 

  15. Knott, A. B., Perkins, G., Schwarzenbacher, R. & Bossy-Wetzel, E. Mitochondrial fragmentation in neurodegeneration. Nat. Rev. Neurosci.9, 505–518 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Qi, X., Qvit, N., Su, Y. C. & Mochly-Rosen, D. A novel Drp1 inhibitor diminishes aberrant mitochondrial fission and neurotoxicity. J. Cell Sci.126, 789–802 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Grohm, J. et al. Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ.19, 1446–1458 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Joshi, A. U., Saw, N. L., Shamloo, M. & Mochly-Rosen, D. Drp1/Fis1 interaction mediates mitochondrial dysfunction, bioenergetic failure and cognitive decline in Alzheimer’s disease. Oncotarget9, 6128–6143 (2018).

    Article  PubMed  Google Scholar 

  19. Disatnik, M. H. et al. Potential biomarkers to follow the progression and treatment response of Huntington’s disease. J. Exp. Med.213, 2655–2669 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Guo, X. et al. Inhibition of mitochondrial fragmentation diminishes Huntington’s disease-associated neurodegeneration. J. Clin. Invest.123, 5371–5388 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mishra, P. & Chan, D. C. Metabolic regulation of mitochondrial dynamics. J. Cell Biol.212, 379–387 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Crotti, A. et al. Mutant Huntingtin promotes autonomous microglia activation via myeloid lineage-determining factors. Nat. Neurosci.17, 513–521 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sarkar, S. et al. Mitochondrial impairment in microglia amplifies NLRP3 inflammasome proinflammatory signaling in cell culture and animal models of Parkinson’s disease. NPJ Parkinsons Dis.3, 30 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Liu, Y., Hao, W., Dawson, A., Liu, S. & Fassbender, K. Expression of amyotrophic lateral sclerosis-linked SOD1 mutant increases the neurotoxic potential of microglia via TLR2. J. Biol. Chem.284, 3691–3699 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Sondag, C. M., Dhawan, G. & Combs, C. K. Beta amyloid oligomers and fibrils stimulate differential activation of primary microglia. J. Neuroinflamm.6, 1 (2009).

    Article  Google Scholar 

  26. Kornfeld, O. S. et al. Interaction of mitochondrial fission factor with dynamin related protein 1 governs physiological mitochondrial function in vivo. Sci. Rep.8, 14034 (2018).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  27. Falchi, A. M. et al. Astrocytes shed large membrane vesicles that contain mitochondria, lipid droplets and ATP. Histochem. Cell Biol.139, 221–231 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Gogvadze, V., Orrenius, S. & Zhivotovsky, B. Multiple pathways of cytochrome c release from mitochondria in apoptosis. Biochim. Biophys. Acta1757, 639–647 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Ryan, K. J. et al. A human microglia-like cellular model for assessing the effects of neurodegenerative disease gene variants. Sci. Transl Med.9, eaai7635 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Cassidy-Stone, A. et al. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev. Cell14, 193–204 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yates, D. Neurodegenerative disease: factoring in astrocytes. Nat. Rev. Neurosci.16, 67 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. West, A. P. et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature520, 553–557 (2015).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  33. Zhong, Z. et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature560, 198–203 (2018).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  34. Chou, S. H. et al. Extracellular mitochondria in cerebrospinal fluid and neurological recovery after subarachnoid hemorrhage. Stroke48, 2231–2237 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zussman, B., Weiner, G. & Ducruet, A. Mitochondrial transfer into the cerebrospinal fluid in the setting of subarachnoid hemorrhage. Neurosurgery82, N11–N13 (2018).

    Article  PubMed  Google Scholar 

  36. Hayakawa, K. et al. Protective effects of endothelial progenitor cell-derived extracellular mitochondria in brain endothelium. Stem Cells36, 1404–1410 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Hayakawa, K. et al. Extracellular mitochondria for therapy and diagnosis in acute central nervous system injury. JAMA Neurol.75, 119–122 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Wilkins, H. M. et al. Extracellular mitochondria and mitochondrial components act as damage-associated molecular pattern molecules in the mouse brain. J. Neuroimmune Pharm.11, 622–628 (2016).

    Article  Google Scholar 

  39. Labzin, L. I., Heneka, M. T. & Latz, E. Innate immunity and neurodegeneration. Annu. Rev. Med69, 437–449 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Glass, C. K., Saijo, K., Winner, B., Marchetto, M. C. & Gage, F. H. Mechanisms underlying inflammation in neurodegeneration. Cell140, 918–934 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. van Niel, G., D’Angelo, G. & Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol.19, 213–228 (2018).

    Article  PubMed  Google Scholar 

  42. Lai, R. C. et al. MSC secretes at least 3 EV types each with a unique permutation of membrane lipid, protein and RNA. J. Extra. Vesicles5, 29828 (2016).

    Article  Google Scholar 

  43. Taylor, A. R., Robinson, M. B., Gifondorwa, D. J., Tytell, M. & Milligan, C. E. Regulation of heat shock protein 70 release in astrocytes: role of signaling kinases. Dev. Neurobiol.67, 1815–1829 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Hooper, C. et al. Wnt3a induces exosome secretion from primary cultured rat microglia. BMC Neurosci.13, 144 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Glebov, K. et al. Serotonin stimulates secretion of exosomes from microglia cells. Glia63, 626–634 (2015).

    Article  PubMed  Google Scholar 

  46. Hessvik, N. P. & Llorente, A. Current knowledge on exosome biogenesis and release. Cell Mol. Life Sci.75, 193–208 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Islam, M. N. et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med.18, 759–765 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Stevens, J. C. et al. Modification of superoxide dismutase 1 (SOD1) properties by a GFP tag—implications for research into amyotrophic lateral sclerosis (ALS). PLoS One5, e9541 (2010).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  49. Minhas, P. S. et al. Macrophage de novo NAD+ synthesis specifies immune function in aging and inflammation. Nat. Immunol.20, 50–63 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Valente, A. J., Maddalena, L. A., Robb, E. L., Moradi, F. & Stuart, J. A. A simple ImageJ macro tool for analyzing mitochondrial network morphology in mammalian cell culture. Acta Histochem.119, 315–326 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

In memory of Ben Barres, whose advice and research inspired this study. This work was supported, in part, by a Stanford Discovery Innovation Award and R01 HL52141 to D.M.-R., a Paul & Daisy Soros Fellowship to P.S.M., a postdoctoral fellowship from the Australian National Health and Medical Research Council (GNT1052961) and the Glenn Foundation Glenn Award to S.A.L., RO1AG058047 to K.I.A., and R35 HL135736 to G.W.D. The authors thank N. Raju and S. Avolicino for assistance with immunohistochemistry and J. Perrino for technical support with electron microscopy.

Author information

Authors and Affiliations

Authors

Contributions

A.U.J. and D.M.-R. generated the hypothesis and the experimental design. A.U.J., D.M.-R. and G.W.D. contributed to the manuscript preparation. K.I.A. contributed to the experimental design. A.U.J. performed isolations of neurons, microglia and astrocytes, collected, analyzed and interpreted the results from earlier mouse studies, and qPCR, ELISA, immunoblots and mitochondrial assays. P.S.M. performed isolations of human monocyte-derived microglia, neurons, microglia and astrocytes, collected data from Seahorse experiments and helped with data analyses. B.H. performed the mitochondrial assays, ELISA and immunoblots and helped with data analyses. S.A.L. isolated microglia and performed microfluidic qPCR and helped with data analyses. All the authors reviewed and edited the manuscript.

Corresponding author

Correspondence to Daria Mochly-Rosen.

Ethics declarations

Competing interests

Patents on P110 and its utility in HD, ALS and other neurodegenerative diseases have been filed by D.M.‐R. and A.U.J. The other authors declare no competing interests.

Additional information

Peer review informationNature Neuroscience thanks Robert Friedlander, Krista Spiller and the other, anonymous, reviewer(s) 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.

Integrated supplementary information

Supplementary Figure 1 Inhibition of excessive mitochondrial fission by treatment with P110 in vivo reduces sustained microglial-astrocytic activation and subsequent pro-inflammatory response in three mouse models of neurodegenerative diseases, AD, HD and ALS.

(a) Area occupied by GFAP immunoreactive astrocytes (brown) was quantified in hippocampus, striatum and spinal cord, in 5xFAD AD mice, in R6/2 HD mice and SOD1 G93A ALS mice, respectively, vehicle- or P110-treated with 3 mg/kg/day for the time indicated in (1a). (b) Area occupied by Iba-1 immunoreactive microglia (brown) was quantified in hippocampus, striatum and spinal cord, in 5xFAD AD mice, in R6/2 HD mice and SOD1 G93A ALS mice, respectively, vehicle- or P110-treated with 3 mg/kg/day for the time indicated in (1a; red rectangles). (n = 5 per group/ 2 sections per mice.). (c) Representative western blot staining for glial fibrillary acidic protein (GFAP), a marker of astrocyte activation, Iba-1, a marker of microglial activation, NLRP3, a marker of inflammasome activation and COX-2, a marker of canonical inflammation in whole brain R6/2 HD mice at endpoint. (d) Representative western blot staining for glial fibrillary acidic protein (GFAP), a marker of astrocyte activation, Iba-1, a marker of microglial activation, NLRP3, a marker of inflammasome activation and COX-2, a marker of canonical inflammation in spinal cord SOD1 G93A mice at endpoint. (e) Representative western blot staining for glial fibrillary acidic protein (GFAP), a marker of astrocyte activation, Iba-1, a marker of microglial activation, NLRP3, a marker of inflammasome activation and COX-2, a marker of canonical inflammation in whole brain lysate in 5XFAD mice at endpoint. 1a. (n = 5 per group). β-actin was used as a loading control. (f) NLRP3 and (g) COX-2 protein levels (determined by quantitating immunostaining) in each mouse model, presented as ratio to the corresponding WT mice. Data were evaluated by one-way ANOVA and Tukey’s multiple comparisons test for multiple testing between each treatment group. All graphs represent mean ± s.d.

Supplementary Figure 2 Excessive mitochondrial fission and dysfunction linked with inflammasome activation in microglia.

(a) Schematic of the experimental design using primary rat microglia treated with LPS (10 ng/ml) for 3 hours, followed by nigericin (1.2µM)-treatment in the presence/ absence of P110 (1 µM added 15 minutes prior to LPS) for 21 hours in serum and antibiotic free DMEM (top). Cellular levels of phospho-p44/42, phosphorylated NFκB and phosphorylated c-Jun N-terminal kinase (JNK) were determined by immunoblotting, quantified and presented as fold-change of control in rat primary microglia; (n=3). (b) Levels of Il-1β and caspase1-p20 were determined in cultured media of these primary microglia by immunoblotting. β-actin in the cells from the corresponding cultures was used as a cell loading control; (n = 2.) (c) Mitochondrial NLRP3 and ASC levels were determined by immunoblotting of mitochondrial fraction, quantified and presented as fold-change of control in these primary microglia. Data were evaluated by one-way ANOVA and Holm-Sidak’s multiple comparisons test for multiple testing between each treatment group; (n=5) All graphs represent mean ± s.d.

Supplementary Figure 3 Transfer of conditioned media of primary rat microglia activates primary rat astrocytes in vitro in a process dependent on Drp1/Fis1 interaction in the microglia and results in Drp1 activation and mitochondrial dysfunction in the astrocytes.

(a) The protocol of cell treatments and media transfer (above). Levels of GFAP protein were determined in cell extracts by immunoblotting rat primary astrocytes treated with supernatant from activated rat primary microglia (MCM); β-actin was used as a loading control; n = 4 (b) IL-1α, IL-1β, TNFα in the culture media of the astrocytes were measured by ELISA; n = 6 and (c) C1q RNA levels in cell extracts were measured by qPCR in cells treated as described in a; n = 5 (d) Drp1 phosphorylation in astrocytes (as a measure of Drp1 activation) were determined by immunoblotting in primary astrocytes treated with supernatant from activated primary microglia; β-actin was used as a loading control; n=4. (e) Mitochondrial levels of NLRP3 and ASC were determined by immunoblotting in the primary astrocytes; β-actin was used as a loading control. Protein levels were quantified and are presented as fold-change of control; n = 3-6. (f) Quantitative PCR analysis of astrocyte-specific phagocytic receptors (multiple EGF-like domains 10, Megf10, and MER proto-oncogene, tyrosine kinase, Mertk) and bridging molecules (receptor tyrosine kinase, Axl, and its ligand, growth arrest-specific 6, Gas6) in primary astrocytes treated with conditioned media, as in a; n=4. (g) OXPHOS in primary astrocytes treated with conditioned media, as in a; n=4. Data were evaluated by one-way ANOVA and Holm-Sidak’s multiple comparisons test for multiple testing between each treatment group. All graphs represent mean ± s.d.

Supplementary Figure 4 Functional mitochondria expelled from astrocytes decrease neuronal death.

(a) Primary cortical rat neurons were treated with conditioned media of primary rat microglia treated as in described in Fig. 2l for 24 hours without (Con) or with LPS/Nig without (Veh) or with P110 (1 μM) and assessed for survival, using LDH release from the neurons. (n=12) (b) Protocol of injury propagation from primary rat microglia activated by LPS to primary cortical rat neurons by media from activated astrocytes (aACM), as in described in Fig. 7a. Where indicated, mitochondria were from the astrocytic media (aACM; filtration through 0.2-μm filters; MitoΔ) and neuronal cell death was determined using LDH release from the neurons. Removal of mitochondria from aACM decreased neuronal survival, indicating the neuroprotective role of functional extracellular mitochondria; (n=10). (c) Primary cortical rat neurons were treated with conditioned media of primary rat astrocytes treated as in described in Fig. 6j for 24 hours. Neuronal cell death was determined using LDH release from the neurons; n=5). (d) Neuroinflammation response propagated from activated microglia to astrocytes to cause cell death of naïve neurons in models of neurodegenerative diseases via expelled and damaged glial mitochondria. Both functional and dysfunctional mitochondria are expelled from both astrocytes and microglia cells. However, mitochondrial dysfunction in microglia triggers the release of more dysfunctional mitochondria relative to functional mitochondria, and these dysfunctional extracellular mitochondria propagate the injurious signal to neurons directly, as well as through inducing mitochondrial dysfunction, activation and neuroinflammation in the astrocytes (left). Inhibiting Drp1/Fis1-mediated pathological fission in the microglia, using a selective inhibitor, such as P110, greatly reduces neuroinflammation, propagation of mitochondrial dysfunction and neuronal cell death. Blocking Drp1Fis1-mediated mitochondrial fission in the microglia increased the transfer benefit to subsequent cell types, in part, by releasing healthier mitochondria into the neuronal milieu (right). Whether the neurons uptake the extracellular mitochondria remains to be determined. Data were evaluated by one-way ANOVA and Holm-Sidak’s multiple comparisons test for multiple testing between each treatment group. All graphs represent mean ± s.d.

Supplementary Figure 5 Image processing for mitochondrial structure analysis.

Images acquired were processed using a macro in Fiji (Image J).

Supplementary Figure 6

Source Data for Western Blots.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Joshi, A.U., Minhas, P.S., Liddelow, S.A. et al. Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration. Nat Neurosci 22, 1635–1648 (2019). https://doi.org/10.1038/s41593-019-0486-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0486-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research