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:

Depletion of microglia and inhibition of exosome synthesis halt tau propagation

Abstract

Accumulation of pathological tau protein is a major hallmark of Alzheimer's disease. Tau protein spreads from the entorhinal cortex to the hippocampal region early in the disease. Microglia, the primary phagocytes in the brain, are positively correlated with tau pathology, but their involvement in tau propagation is unknown. We developed an adeno-associated virus–based model exhibiting rapid tau propagation from the entorhinal cortex to the dentate gyrus in 4 weeks. We found that depleting microglia dramatically suppressed the propagation of tau and reduced excitability in the dentate gyrus in this mouse model. Moreover, we demonstrate that microglia spread tau via exosome secretion, and inhibiting exosome synthesis significantly reduced tau propagation in vitro and in vivo. These data suggest that microglia and exosomes contribute to the progression of tauopathy and that the exosome secretion pathway may be a therapeutic target.

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

Figure 1: Tau propagates to the DG after the injection of AAV-GFP/tau in the MEC of the mouse brain.
Figure 2: Immunofluorescence for tau with cellular markers.
Figure 3: Microglial depletion suppresses tau propagation in the DG of AAV-GFP/tau-injected mice and PS19 tau mice.
Figure 4: Reduced population spike responses in AAV-GFP/tau mice are rescued by depletion of microglia.
Figure 5: Microglia phagocytose and secrete human tau in exosomes, which facilitates tau propagation to neurons in vitro and in vivo.
Figure 6: Characterization of tau-containing extracellular vesicles from microglia depleted PS19 mouse brain.
Figure 7: nSMase2 silencing or inhibition reduces exosomal secretion and exosome-mediated tau transmission from microglia to neurons.
Figure 8: Inhibition of nSMase2 suppresses tau propagation in the DG of AAV-GFP/tau-injected and PS19 tau mice.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  2. Clavaguera, F. et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell Biol. 11, 909–913 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ahmed, Z. et al. A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity. Acta Neuropathol. 127, 667–683 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Boluda, S. et al. Differential induction and spread of tau pathology in young PS19 tau transgenic mice following intracerebral injections of pathological tau from Alzheimer's disease or corticobasal degeneration brains. Acta Neuropathol. 129, 221–237 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Iba, M. et al. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer's-like tauopathy. J. Neurosci. 33, 1024–1037 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Harris, J.A. et al. Human P301L-mutant tau expression in mouse entorhinal-hippocampal network causes tau aggregation and presynaptic pathology but no cognitive deficits. PLoS ONE 7, e45881 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. de Calignon, A. et al. Propagation of tau pathology in a model of early Alzheimer's disease. Neuron 73, 685–697 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Liu, L. et al. Trans-synaptic spread of tau pathology in vivo. PLoS ONE 7, e31302 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Peeraer, E. et al. Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice. Neurobiol. Dis. 73, 83–95 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Yamada, K. et al. Neuronal activity regulates extracellular tau in vivo. J. Exp. Med. 211, 387–393 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sanders, D.W. et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82, 1271–1288 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Vingtdeux, V., Sergeant, N. & Buee, L. Potential contribution of exosomes to the prion-like propagation of lesions in Alzheimer's disease. Front. Physiol. 3, 229 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Saman, S. et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J. Biol. Chem. 287, 3842–3849 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Fiandaca, M.S. et al. Identification of preclinical Alzheimer's disease by a profile of pathogenic proteins in neurally derived blood exosomes: a case-control study. Alzheimers Dement. 11, 600–607 (2015).

    Article  PubMed  Google Scholar 

  15. Neumann, H., Kotter, M.R. & Franklin, R.J. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain 132, 288–295 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Schafer, D.P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. El Andaloussi, S., Mager, I., Breakefield, X.O. & Wood, M.J. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347–357 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Ghoshal, N. et al. Tau-66: evidence for a novel tau conformation in Alzheimer's disease. J. Neurochem. 77, 1372–1385 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Serrano-Pozo, A. et al. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer's disease. Am. J. Pathol. 179, 1373–1384 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Shevtsova, Z., Malik, J.M., Michel, U., Bahr, M. & Kugler, S. Promoters and serotypes: targeting of adeno-associated virus vectors for gene transfer in the rat central nervous system in vitro and in vivo. Exp. Physiol. 90, 53–59 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Schofield, E., Kersaitis, C., Shepherd, C.E., Kril, J.J. & Halliday, G.M. Severity of gliosis in Pick's disease and frontotemporal lobar degeneration: tau-positive glia differentiate these disorders. Brain 126, 827–840 (2003).

    Article  PubMed  Google Scholar 

  22. Fricker, M. et al. MFG-E8 mediates primary phagocytosis of viable neurons during neuroinflammation. J. Neurosci. 32, 2657–2666 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yoshiyama, Y. et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53, 337–351 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Elmore, M.R. et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380–397 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Qu, Y., Franchi, L., Nunez, G. & Dubyak, G.R. Nonclassical IL-1 beta secretion stimulated by P2X7 receptors is dependent on inflammasome activation and correlated with exosome release in murine macrophages. J. Immunol. 179, 1913–1925 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Kadiu, I., Narayanasamy, P., Dash, P.K., Zhang, W. & Gendelman, H.E. Biochemical and biologic characterization of exosomes and microvesicles as facilitators of HIV-1 infection in macrophages. J. Immunol. 189, 744–754 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Perez-Gonzalez, R., Gauthier, S.A., Kumar, A. & Levy, E. The exosome secretory pathway transports amyloid precursor protein carboxyl-terminal fragments from the cell into the brain extracellular space. J. Biol. Chem. 287, 43108–43115 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Thery, C., Amigorena, S., Raposo, G. & Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. Ch. 3, Unit 3.22 (2006).

  29. Trajkovic, K. et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319, 1244–1247 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Brown, G.C. & Neher, J.J. Microglial phagocytosis of live neurons. Nat. Rev. Neurosci. 15, 209–216 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Ittner, L.M. et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models. Cell 142, 387–397 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Clavaguera, F. et al. Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proc. Natl. Acad. Sci. USA 110, 9535–9540 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lasagna-Reeves, C.A. et al. Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci. Rep. 2, 700 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Agosta, F. et al. Myeloid microvesicles in cerebrospinal fluid are associated with myelin damage and neuronal loss in mild cognitive impairment and Alzheimer disease. Ann. Neurol. 76, 813–825 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Lasagna-Reeves, C.A. et al. Identification of oligomers at early stages of tau aggregation in Alzheimer's disease. FASEB J. 26, 1946–1959 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Théry, C., Ostrowski, M. & Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 9, 581–593 (2009).

    Article  PubMed  CAS  Google Scholar 

  37. Lu, Z. et al. Phagocytic activity of neuronal progenitors regulates adult neurogenesis. Nat. Cell Biol. 13, 1076–1083 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Santacruz, K. et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science 309, 476–481 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Castillo-Carranza, D.L. et al. Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J. Neurosci. 34, 4260–4272 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Danzer, K.M. et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 7, 42 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Alvarez-Erviti, L. et al. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol. Dis. 42, 360–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Arellano-Anaya, Z.E. et al. Prion strains are differentially released through the exosomal pathway. Cell. Mol. Life Sci. 72, 1185–1196 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Phinney, A.L. et al. Cerebral amyloid induces aberrant axonal sprouting and ectopic terminal formation in amyloid precursor protein transgenic mice. J. Neurosci. 19, 8552–8559 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yamamoto, M., Kiyota, T., Walsh, S.M. & Ikezu, T. Kinetic analysis of aggregated amyloid-beta peptide clearance in adult bone-marrow-derived macrophages from APP and CCL2 transgenic mice. J. Neuroimmune Pharmacol. 2, 213–221 (2007).

    Article  PubMed  Google Scholar 

  45. Lewis, J. et al. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat. Genet. 25, 402–405 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Jicha, G.A., Bowser, R., Kazam, I.G. & Davies, P. Alz-50 and MC-1, a new monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau. J. Neurosci. Res. 48, 128–132 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Otvos, L. Jr. et al. Monoclonal antibody PHF-1 recognizes tau protein phosphorylated at serine residues 396 and 404. J. Neurosci. Res. 39, 669–673 (1994).

    Article  CAS  PubMed  Google Scholar 

  48. Butovsky, O. et al. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann. Neurol. 77, 75–99 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Butovsky, O. et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci. 17, 131–143 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Vosshall, L.B., Wong, A.M. & Axel, R. An olfactory sensory map in the fly brain. Cell 102, 147–159 (2000).

    Article  CAS  PubMed  Google Scholar 

  51. Hama, H. et al. Scale: a chemical approach for fluorescence imaging and reconstruction of transparent mouse brain. Nat. Neurosci. 14, 1481–1488 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Tyler, W.A. & Haydar, T.F. Multiplex genetic fate mapping reveals a novel route of neocortical neurogenesis, which is altered in the Ts65Dn mouse model of Down syndrome. J. Neurosci. 33, 5106–5119 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kiyota, T. et al. CCL2 accelerates microglia-mediated Aβ oligomer formation and progression of neurocognitive dysfunction. PLoS ONE 4, e6197 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Kiyota, T., Ingraham, K.L., Jacobsen, M.T., Xiong, H. & Ikezu, T. FGF2 gene transfer restores hippocampal functions in mouse models of Alzheimer's disease and has therapeutic implications for neurocognitive disorders. Proc. Natl. Acad. Sci. USA 108, E1339–E1348 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kiyota, T. et al. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP+PS1 mice. Gene Ther. 19, 724–733 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Beaudoin, G.M. III et al. Culturing pyramidal neurons from the early postnatal mouse hippocampus and cortex. Nat. Protoc. 7, 1741–1754 (2012).

    Article  CAS  PubMed  Google Scholar 

  57. Flach, K. et al. Tau oligomers impair artificial membrane integrity and cellular viability. J. Biol. Chem. 287, 43223–43233 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Blasi, E., Barluzzi, R., Bocchini, V., Mazzolla, R. & Bistoni, F. Immortalization of murine microglial cells by a v-raf/v-myc carrying retrovirus. J. Neuroimmunol. 27, 229–237 (1990).

    Article  CAS  PubMed  Google Scholar 

  59. Ellman, G.L., Courtney, K.D., Andres, V. Jr. & Feather-Stone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 7, 88–95 (1961).

    Article  CAS  PubMed  Google Scholar 

  60. Roccaro, A.M. et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Invest. 123, 1542–1555 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Freilich, R.W., Woodbury, M.E. & Ikezu, T. Integrated expression profiles of mRNA and miRNA in polarized primary murine microglia. PLoS ONE 8, e79416 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Tabatadze, N. et al. Inhibition of neutral sphingomyelinase-2 perturbs brain sphingolipid balance and spatial memory in mice. J. Neurosci. Res. 88, 2940–2951 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Ericsson (Electron Microscopy Facility, Harvard Medical School) for electron microscopic imaging services; P. Davies (Albert Einstein College of Medicine) for providing the MC1, CP13 and PHF1 antibodies; R. Kayed (University of Texas Medical Branch) for providing the T22 polyclonal antibody; Plexxikon, Inc. for providing PLX3397 and control chows; and M. Hasselmo and S. Przedborski for critical reading of the manuscript. This work is supported in part by grants from Alzheimer's Association (T.I.), Alzheimer's Art Quilt Initiative (T.I.), Boston University Alzheimer's Disease Center (P30AG013846, T.I.), BrightFocus Foundation (H.A.) and Coins for Alzheimer's Research Trust (T.I.).

Author information

Authors and Affiliations

Authors

Contributions

T.I. and H.A. designed the AAV-GFP/tau injection mouse model and initiated the study. M.M. and J.L. performed field recording electrophysiology and analyses; S.I. and H.A. cultured primary neuronal cells; H.A., S.I. and T.I. purified exosomes for electron microscopy and biochemical analyses; T.H. performed two-photon imaging and laser-scanning confocal microscopy; O.B. generated and provided the Pr2ry12 antibody, performed data analysis and edited the manuscript; B.W. provided tau antibodies, performed data analysis and wrote the manuscript; S.K. generated and provided AAV vectors, performed data analysis and edited the manuscript; and H.A., S.I., J.L., S.T., M.M. and T.I. designed and performed experiments and data analyses and wrote the manuscript.

Corresponding author

Correspondence to Tsuneya Ikezu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 AAV-GFP injection in the MEC of the mouse brain

C57Bl/6 mice at 4 months of age were injected with AAV-GFP into the MEC and sacrificed at 7 days post injection (dpi). (a) Brains were sectioned with 0.5-mm thickness, followed by 4M urea containing Scale solution treatment to make brain transparent. (c) Green fluorescence protein (GFP) was detected in MEC injected site. (d) Immunofluorescence of GFP (green) with GFAP (astrocytic marker, red) (e) Iba1 (mononuclear phagocyte maker, red) and (f) NeuN (neuron marker, red) in the MEC of injected mouse brain; Scale bars: 25 μm. (Scheme in b adapted from The Mouse Brain in Stereotaxic Coordinates 2nd ed (2001); p.333).

Supplementary Figure 2 Cytotoxic changes of tau-bearing neurons in the DG and reduction of changes by microglial depletion or nSMase2 inhibition in two different tau mouse models

(a) Cleaved caspase-3 (green), AT8 (pTau, red), and Dapi (blue) staining in the dentate granule cell layer (GCL) of AAV-GFP/tau mice at 28 dpi. Scale bar: 20 μm. GFP signal was quenched by methanol/acetone treatment of tissue sections prior to the immunostaining. (b,c) AAV-GFP/tau mice or PS19 mice were treated for microglial depletion by feeding mice with PLX3397 chow (upper panel) or nSMase2 inhibition by ip injection of GW4869 (lower panel). The frozen tissue sections were immunostained for cleaved caspase-3 (red) and Dapi (blue) in the GCL of AAV-GFP/tau mice at 28 dpi (A) or PS19 tau mice at 3.5 months of age (B); scale bars: 10 μm.

Supplementary Figure 3 Microglial density after AAV-GFP/tau injection into the MEC and depletion of microglia by clodronate liposome or PLX3397

(a,b) AAV-GFP or AAV-GFP/tau mice are subjected to immunofluorescence of Iba1 (mononuclear phagocyte marker, red) and Dapi (blue) in the DG at 7 or 28 dpi. (b) Quantification of microglial density in the DG. N.D.: No statistical difference between AAV-GFP and AAV-GFP/tau mice at 7 or 28 dpi as determined by one-way ANOVA (p>0.999, t(32)=0.0, n=3, 17 sections per group for 7 dpi; p=0.3231, t(42)=0.999, n=3, 22 sections mice per group for 28 dpi). Scale bar: 50 μm (c) C57BL/6 mice are pre-treated with clodronate liposome (CL-Lip) or PBS liposome (PBS-Lip) through a cannula implanted into the lateral ventricle at the time of AAV-GFP/tau injection until 28 dpi (a total of 4 week treatment, see Supplementary Figure 6 for details). PBS-Lip was injected in the control group. For PLX3397 study, C57BL/6 mice are pre-fed with the control or PLX-containing chow for 4 weeks prior to the AAV-GFP/tau injection, and maintained in the same chow until 28 dpi (a total of 8 week treatment). immunofluorescence of P2ry12 (microglia-specific marker, green) and CD169 (infiltrating monocytes marker, red) in the DG of AAV-GFP/tau mice at 28 dpi. Scale bar: 50 μm.

Supplementary Figure 4 Colocalization of pTau within Iba1-positive microglia

(a) Sequential imaging of confocal microscopy in the hippocampal area of the 6 month-old PS19 mouse brain. Green; AT8 (red), Iba1 (green). Scale bar: 10 μm (b) Double-immunogold labeling and electron microscopy in the hippocampal area of the 9-month-old PS19 mouse brain. 15-nm gold particles bind to Iba1 and 10-nm gold particles bind to PHF1 (pTau at pSer396/Ser404). Scale bars: 100 nm.

Supplementary Figure 5 Microglial depletion by clodronate liposome

(a) Cannula coordinate and time point for the CL-liposome ICV injection. (b) CL or PBS-Lip was ICV injected once a week for 4 weeks through the implanted cannula for one month after the AAV-GFP/tau injection. (c) Iba1 and Dapi staining in periventricular zone (top panels) and hippocampus (bottom panels). Scale bars: top panels: 25 μm; bottom panels: 50 μm (Scheme in a adapted from The Mouse Brain in Stereotaxic Coordinates second edition (2001), p. 83).

Supplementary Figure 6 Quantification of hTau in the MEC of AAV-GFP/tau-injected mice and PLX treatment and gene expression profile of immune molecules in the hippocampus

PLX3397 (or control) chow-fed C57BL/6 mice were injected with AAV-GFP/tau into the MEC and hippocampal regions were isolated at 28 dpi. (a) Quantification of hTau in the MEC using hTau specific ELISA. No statistical difference between control- and PLX-chow fed groups (p=0.8515, t(4)=0.1996, n=3 per group). (b) Gene expression profiles of pro-inflammatory cytokines (Tnfa, Il1b, and Il6) and anti-inflammatory cytokines (Il10 and Tgfb1) in the hippocampus of control or PLX chow-fed mice with AAV-GFP/tau injection. Each column represents ΔΔCT value vs. age-matched untreated C57BL/6 mice. p<0.0001, F(1,20)=77.29, n= 3 per group as determined by two-way ANOVA and multiple comparison tests. t(20)=2.987 (Tnfa), 2.877 (Il1b), 5.422 (Il6), 4.852 (Il10), and 3.610 (Tgfb1). *P < 0.05, **P < 0.01 and ***P < 0.001 between control and PLX group as determined by two-way ANOVA and multiple comparison tests (n=3 per group).

Supplementary Figure 7 Aggregation of purified hTau and microglial phagocytosis in vitro.

(a) Recombinant hTau protein was aggregated in the presence of heparin for 0-24 hrs as described in methods, and subjected to western blotting using HT7 (anti-hTau mAb). High molecular weight tau bands (120kDa and above) are visible even at 0 time point. (b) Quantification of tau oligomers by dot-blot analysis of aggregated tau protein for 0-72 hours. Tau oligomerization is increased after 3 hr incubation and is saturated by 24 hrs. (c) Murine primary microglia (MG) are treated with aggregated tau ± 1μM cytochalasin D (CyD, phagocytosis inhibitor) for 30 min, and the cells were washed with PBS and fixed with 4% paraformaldehyde for immunofluorescence using HT7 (green) and Dapi (blue); scale bar: 50μm. (d) After the phagocytosis assay of MG or BV-2 cells (murine microglial cell line), the cells were trypsinized to remove extracellular tau, and subjected to hTau ELISA. MG: p=0.0005, F(2,6)=34.93, q(6)=11.82 (MG no hTau vs. MG hTau), 5.781 (MG no hTau vs. MG hTau+CyD), and 6.039 (MG hTau vs. MG hTau+CyD). BV2: p=0.0005, F(2,6)=34.94, q(6)=11.80 (BV2 no hTau vs. BV2 hTau), 5.285 (BV2 no hTau vs. BV2 hTau+CyD), 6.517 (BV2 hTau vs. BV2+CyD). *P < 0.05, **P < 0.01 and ***P < 0.001 as determined by one-way ANOVA and Tukey post hoc.

Supplementary Figure 8 nSMase2 knockdown and purified tau-containing exosomes injected into the OML of mouse brain

(a) Neutral sphingomyeliase-2 (nSMase2) mRNA expression levels in mouse primary microglia treated with siRNA against murine nSMase2 or scramble siRNA. (b) Purified tau-containing exosomes from hTau-phagocytosed microglia was DiI-labeled. (c) One μl of the exosomal fraction from microglia with hTau+LPS+ATP+ or hTau-LPS-ATP+ treatment, and 1 to 50 ng/μl of aggregated recombinant hTau were blotted with HT7 or Tsg101. Full-length blots/gels are presented in Supplementary Figure 10. (d) Immunofluorescence of of HT7 (green), DiI (exosome, red), Dapi (blue) of mouse brain sections after injection of tau containing exosomes to the OML of the DG of C57BL/6 mice. Scale bar: 200 μm.

Supplementary Figure 9 Quantification of hTau in the MEC of AAV-GFP/tau injected mice and GW4869 treatment and gene expression profile of immune molecules in the hippocampus

C57BL/6 mice were injected with AAV-GFP/tau into the MEC, had daily ip injection of 1.25 mg/kg of GW4869 or 200μl of 5% DMSO in saline (control vehicle) from 0 to 28 dpi, and the MEC and hippocampal regions were isolated at 28 dpi. (a) Quantification of hTau in the MEC using hTau specific ELISA. No statistical difference between DMSO- and GW4869-treated groups (p=0.9295, t(4)=0.0942, n=3 per group). (b) Gene expression profiles of pro-inflammatory cytokines (Tnfa, Il1b, and Il6) and anti-inflammatory cytokines (Il10 and Tgfb1) in the hippocampus of DMSO or GW4869-treated AAV-GFP/tau mice. Each column represents ΔΔCT value vs. age-matched untreated C57BL/6 mice. p<0.0001, F(1,20)=28.89, t(20)=2.819 (Tnfa), 1.535 (Il1b), 1.114 (Il6), 4.725 (Il10), and 1.826 (Tgfb1) groups as determined by two-way ANOVA and multiple comparison tests (n=3 per group). *** P < 0.001 between DMSO and GW4869.

Supplementary Figure 10 Full-length blots for Figures 5 and 7 and Supplementary Figure 8c.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Asai, H., Ikezu, S., Tsunoda, S. et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 18, 1584–1593 (2015). https://doi.org/10.1038/nn.4132

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4132

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