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:

TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration

Abstract

Triggering receptor expressed on myeloid cell 2 (TREM2) is linked to risk of neurodegenerative disease. However, the function of TREM2 in neurodegeneration is still not fully understood. Here, we investigated the role of microglial TREM2 in TAR DNA-binding protein 43 (TDP-43)-related neurodegeneration using virus-mediated and transgenic mouse models. We found that TREM2 deficiency impaired phagocytic clearance of pathological TDP-43 by microglia and enhanced neuronal damage and motor impairments. Mass cytometry analysis revealed that human TDP-43 (hTDP-43) induced a TREM2-dependent subpopulation of microglia with high CD11c expression and phagocytic ability. Using mass spectrometry (MS) and surface plasmon resonance (SPR) analysis, we further demonstrated an interaction between TDP-43 and TREM2 in vitro and in vivo as well as in human tissues from individuals with amyotrophic lateral sclerosis (ALS). We computationally identified regions within hTDP-43 that interact with TREM2. Our data highlight that TDP-43 is a possible ligand for microglial TREM2 and that this interaction mediates neuroprotection of microglia in TDP-43-related neurodegeneration.

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: TREM2 deficiency aggravates hTDP-43-induced behavioral deficits and neurodegeneration.
Fig. 2: TREM2 deficiency abolishes hTDP-43-induced CD11c+ microglia subpopulation.
Fig. 3: TREM2 deficiency locks microglia into a homeostatic state in TDP-43-induced neurodegeneration.
Fig. 4: TREM2 deficiency impairs microglial phagocytosis of pathological hTDP-43 protein.
Fig. 5: TREM2 deficiency leads to increased accumulation of pathological hTDP-43 protein.
Fig. 6: hTDP-43 interacts with TREM2 in vitro and in vivo in the mouse brain.
Fig. 7: hTDP-43 interacts with TREM2 in human tissues and in silico.

Similar content being viewed by others

Data availability

The MS proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE63 partner repository with the accession number PXD029429 and 10.6019/PXD029429. Other information that supports the findings of this study is available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Hickman, S., Izzy, S., Sen, P., Morsett, L. & El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 21, 1359–1369 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ulland, T. K. & Colonna, M. TREM2—a key player in microglial biology and Alzheimer disease. Nat. Rev. Neurol. 14, 667–675 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Guerreiro, R. et al. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 368, 117–127 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Jonsson, T. et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 368, 107–116 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Wang, Y. et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell 160, 1061–1071 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhao, Y. et al. TREM2 is a receptor for β-amyloid that mediates microglial function. Neuron 97, 1023–1031 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ou, S. H., Wu, F., Harrich, D., Garcia-Martinez, L. F. & Gaynor, R. B. Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. J. Virol. 69, 3584–3596 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Neumann, M. et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314, 130–133 (2006).

    Article  CAS  PubMed  Google Scholar 

  9. Cairns, N. J. et al. TDP-43 in familial and sporadic frontotemporal lobar degeneration with ubiquitin inclusions. Am. J. Pathol. 171, 227–240 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cady, J. et al. TREM2 variant p.R47H as a risk factor for sporadic amyotrophic lateral sclerosis. JAMA Neurol. 71, 449–453 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Rayaprolu, S. et al. TREM2 in neurodegeneration: evidence for association of the p.R47H variant with frontotemporal dementia and Parkinson’s disease. Mol. Neurodegener. 8, 19 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Krasemann, S. et al. The TREM2–APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566–581 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Maniatis, S. et al. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science 364, 89–93 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Walker, A. K. et al. Functional recovery in new mouse models of ALS/FTLD after clearance of pathological cytoplasmic TDP-43. Acta Neuropathol. 130, 643–660 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Perry, V. H., Nicoll, J. A. & Holmes, C. Microglia in neurodegenerative disease. Nat. Rev. Neurol. 6, 193–201 (2010).

    Article  PubMed  Google Scholar 

  16. Mrdjen, D. et al. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 48, 380–395 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Eyo, U. B. et al. P2Y12R-dependent translocation mechanisms gate the changing microglial landscape. Cell Rep. 23, 959–966 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Spiller, K. J. et al. Microglia-mediated recovery from ALS-relevant motor neuron degeneration in a mouse model of TDP-43 proteinopathy. Nat. Neurosci. 21, 329–340 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Steinacker, P. et al. TDP-43 in cerebrospinal fluid of patients with frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Arch. Neurol. 65, 1481–1487 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Zhong, J. et al. The interactome of a PTB domain-containing adapter protein, Odin, revealed by SILAC. J. Proteom. 74, 294–303 (2011).

    Article  CAS  Google Scholar 

  22. Guenther, E. L. et al. Atomic structures of TDP-43 LCD segments and insights into reversible or pathogenic aggregation. Nat. Struct. Mol. Biol. 25, 463–471 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lee, E. B., Lee, V. M. & Trojanowski, J. Q. Gains or losses: molecular mechanisms of TDP43-mediated neurodegeneration. Nat. Rev. Neurosci. 13, 38–50 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Li, Q. et al. Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron 101, 207–223 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Liu, Y. U. et al. Neuronal network activity controls microglial process surveillance in awake mice via norepinephrine signaling. Nat. Neurosci. 22, 1771–1781 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Parhizkar, S. et al. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE. Nat. Neurosci. 22, 191–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Leyns, C. E. G. et al. TREM2 function impedes tau seeding in neuritic plaques. Nat. Neurosci. 22, 1217–1222 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lee, C. Y. D. et al. Elevated TREM2 gene dosage reprograms microglia responsivity and ameliorates pathological phenotypes in Alzheimer’s disease models. Neuron 97, 1032–1048 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Konishi, H. & Kiyama, H. Microglial TREM2/DAP12 signaling: a double-edged sword in neural diseases. Front. Cell. Neurosci. 12, 206 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Mazaheri, F. et al. TREM2 deficiency impairs chemotaxis and microglial responses to neuronal injury. EMBO Rep. 18, 1186–1198 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zheng, H. et al. TREM2 promotes microglial survival by activating Wnt/β-catenin pathway. J. Neurosci. 37, 1772–1784 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guan, Z. et al. Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Nat. Neurosci. 19, 94–101 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Gu, N. et al. Spinal microgliosis due to resident microglial proliferation is required for pain hypersensitivity after peripheral nerve injury. Cell Rep. 16, 605–614 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Otero, K. et al. Macrophage colony-stimulating factor induces the proliferation and survival of macrophages via a pathway involving DAP12 and β-catenin. Nat. Immunol. 10, 734–743 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  36. Fu, R., Shen, Q., Xu, P., Luo, J. J. & Tang, Y. Phagocytosis of microglia in the central nervous system diseases. Mol. Neurobiol. 49, 1422–1434 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hong, S. et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Koizumi, S. et al. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature 446, 1091–1095 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Tufail, Y. et al. Phosphatidylserine exposure controls viral innate immune responses by microglia. Neuron 93, 574–586 e578 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Das, R. & Chinnathambi, S. Microglial priming of antigen presentation and adaptive stimulation in Alzheimer’s disease. Cell. Mol. Life Sci. 76, 3681–3694 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Harms, A. S. et al. MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J. Neurosci. 33, 9592–9600 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bulloch, K. et al. CD11c/EYFP transgene illuminates a discrete network of dendritic cells within the embryonic, neonatal, adult, and injured mouse brain. J. Comp. Neurol. 508, 687–710 (2008).

    Article  PubMed  Google Scholar 

  43. Sheean, R. K. et al. Association of regulatory T-cell expansion with progression of amyotrophic lateral sclerosis: a study of humans and a transgenic mouse model. JAMA Neurol. 75, 681–689 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Sato-Hashimoto, M. et al. Microglial SIRPα regulates the emergence of CD11c+ microglia and demyelination damage in white matter. eLife 8, e4202 (2019).

    Article  Google Scholar 

  45. Atagi, Y. et al. Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2). J. Biol. Chem. 290, 26043–26050 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Feiler, M. S. et al. TDP-43 is intercellularly transmitted across axon terminals. J. Cell Biol. 211, 897–911 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhong, L. et al. Soluble TREM2 ameliorates pathological phenotypes by modulating microglial functions in an Alzheimer’s disease model. Nat. Commun. 10, 1365 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Kober, D. L. et al. Neurodegenerative disease mutations in TREM2 reveal a functional surface and distinct loss-of-function mechanisms. eLife 5, e20391 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Yeh, F. L., Wang, Y., Tom, I., Gonzalez, L. C. & Sheng, M. TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-β by microglia. Neuron 91, 328–340 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Ling, J. P., Pletnikova, O., Troncoso, J. C. & Wong, P. C. TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD. Science 349, 650–655 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Clarkson, B. D. S., Patel, M. S., LaFrance-Corey, R. G. & Howe, C. L. Retrograde interferon-γ signaling induces major histocompatibility class I expression in human-induced pluripotent stem cell-derived neurons. Ann. Clin. Transl Neurol. 5, 172–185 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Kim, J.Y., Grunke, S.D., Levites, Y., Golde, T.E. & Jankowsky, J.L. Intracerebroventricular viral injection of the neonatal mouse brain for persistent and widespread neuronal transduction. J. Vis. Exp. https://doi.org/10.3791/51863 (2014).

  53. Peng, J. et al. Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury. Nat. Commun. 7, 12029 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gittins, R. & Harrison, P. J. Neuronal density, size and shape in the human anterior cingulate cortex: a comparison of Nissl and NeuN staining. Brain Res. Bull. 63, 155–160 (2004).

    Article  PubMed  Google Scholar 

  55. Pang, Y. P. FF12MC: a revised AMBER forcefield and new protein simulation protocol. Proteins 84, 1490–1516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Jorgensen, W. L., Chandreskhar, J., Madura, J. D., Impey, R. W. & Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 79, 926–935 (1983).

    Article  CAS  Google Scholar 

  57. Pang, Y.-P. Use of 1–4 interaction scaling factors to control the conformational equilibrium between α-helix and β-strand. Biochem. Biophys. Res. Commun. 457, 183–186 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Larini, L., Mannella, R. & Leporini, D. Langevin stabilization of molecular-dynamics simulations of polymers by means of quasisymplectic algorithms. J. Chem. Phys. 126, 104101 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Darden, T. A., York, D. M. & Pedersen, L. G. Particle mesh Ewald: an N log(N) method for Ewald sums in large systems. J. Chem. Phys. 98, 10089–10092 (1993).

    Article  CAS  Google Scholar 

  60. Pang, Y. P. Low-mass molecular dynamics simulation for configurational sampling enhancement: more evidence and theoretical explanation. Biochem. Biophys. Rep. 4, 126–133 (2015).

    PubMed  PubMed Central  Google Scholar 

  61. Joung, I. S. & Cheatham, T. E. Determination of alkali and halide monovalent ion parameters for use in explicitly solvated biomolecular simulations. J. Phys. Chem. B 112, 9020–9041 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang, J. et al. Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency. Nature 588, 459–465 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Colonna (Washington University) for providing Trem2-KO mice; R.L. Klein (Louisiana State University) for providing AAV virus; V.A. Lennon (Mayo Clinic Rochester) for thoughtful discussion and manuscript editing; W. Rossoll (Mayo Clinic Florida) for insightful suggestions; A.J. Johnson (Mayo Clinic Rochester), Y. Zhu (Mayo Clinic Rochester) and K.D. Pavelko (Mayo Clinic Rochester Immune Monitoring Core) for assistance with CyTOF sample preparation and data analysis; A. Pandey (Mayo Clinic Rochester) and the Mayo Clinic Proteomics Core (a shared resource of the Mayo Clinic Cancer Center) for LC–MS/MS experiment and data analysis; C. Howe and B. Clarkson (Mayo Clinic Rochester) for providing iPSC-derived neurons and R.M. Weinshilboum (Mayo Clinic Rochester) for assistance with biochemistry experiments. The current study is supported by NIH grants R21AG064159 and R01NS088627 to L.-J.W., R01AG066395 to G.B. and N.Z. and U19AG069701 to G.B., Y.A.M., N.Z., C.-C.L., D.W.D. and L.-J.W.

Author information

Authors and Affiliations

Authors

Contributions

M.X., Y.U.L. and L.-J.W. designed the experiments and wrote the manuscript. M.X. and Y.U.L. performed most of the experiments. S.Z., L.Z., U.S., Y.Z., N.Z. and C.-C.L. performed some of the experiments. M.X. and D.B.B. performed data analysis. Y.-P.P. designed, performed and analyzed the MD simulation study. J.Z performed and analyzed the MS data. Y.A.M., L.W., D.W.D., M.P.M. and G.B. provided resources for specific experiments.

Corresponding authors

Correspondence to Yong U. Liu or Long-Jun Wu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review information

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

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 Characterizations of GFP-hTDP-43 expression in a neonatal TDP-43 mouse model.

GFP-hTDP-43 expression was induced via intracerebroventricular injection of AAV9.CAG.hTDP-43.GFP in C57/BL6 (WT) neonatal mice. a, Representative image of brain GFP-hTDP-43 expression in WT mice at 21 days post-infection (dpi). Primary motor cortex (M1), supplementary motor area (M2) and lateral ventricle (LV) are separated by dashed line. Scale bar, 500 µm. Inset shows hTDP-43 expression at higher magnification as indicated by the area in dotted yellow box. Scale bar, 100 µm. b, Representative image of spinal cord (SC) GFP-hTDP-43 expression in WT mice at 21 dpi. Scale bar, 200 µm. c, Representative image (c) and quantification (d, n = 4) of GFP-hTDP-43 expression in the brain regions of cerebral cortex, hippocampus, thalamus and brainstem in WT mice at 21 dpi. Scale bar, 100 μm. e,f, Representative images (e) and quantification (f, n = 7) of co-immunostaining of GFP-hTDP-43 (green) with neuron marker NeuN (red), microglia marker Iba1 (red), astrocyte marker GFAP (red), and oligodendroglia marker CNPase (red) in the primary motor cortex of WT mice at 21 dpi. Scale bar, 100 µm. g, Representative images of p-hTDP-43 expression in the brain regions of hippocampus, thalamus, brainstem at 35 dpi. Scale bar, 20 µm. In d and f, data represented as mean ± SEM.

Extended Data Fig. 2 Characterizations of motor deficits and neuronal loss in a neonatal TDP-43 mouse model.

GFP-hTDP-43 expression was induced via intracerebroventricular injection of AAV9.CAG.hTDP-43.GFP in neonatal mice (AAV9.CAG.GFP as control). a, Body weights were measured separately for male and female of indicated groups at 35 dpi. b,c, Representative traces (b) and quantification (c) of locomotor activity in an open field test of indicated groups at 35 dpi. d,e, Representative immunostaining images (d) and quantification (e) of NeuN (red) with DAPI (blue) in the primary motor cortex of indicated groups at 35 dpi. M1 and M2 are separated by dashed line. Scale bar, 200 µm. Insets show NeuN staining in layer 5 of primary motor cortex at higher magnification as indicated by the area in dotted white box. Scale bar, 20 µm. In a, c, and e, Significance was calculated using Two-way ANOVA, Tukey’s post-hoc analysis. Data represented as mean ± SEM. n.s., not significant, *P < 0.05, **P < 0.01, *** P < 0.001. a, n = 13 per group, female: P < 0.0001, F 3,36 = 17.08; male: P < 0.0001, F 3,36 = 21.72; c, n = 25 per group, P = 0.0008, F 3,66 = 6.339; e, n = 10 per group, P < 0.0001, F 3,27 = 195.1.

Extended Data Fig. 3 TREM2 deficiency leads to increased accumulation of hTDP-43 protein.

hTDP-43 protein expression was induced via intracerebroventricular injection of AAV9.CAG.hTDP-43 in neonatal mouse (AAV9.CAG.Empty as control). a, Schematic representation of the velocity sedimentation protocol for separating soluble from Sarkosyl-insoluble aggregated proteins from an aqueous homogenate of brain tissue. b, Whole brain hTDP-43 and p-hTDP-43 (Ser409/410) immunoblots of the soluble fraction of WT and TREM2 KO mice at 70 dpi. p-hTDP-43 (Ser409/410) antibody detected a ~25 kD fragment in the soluble fractions. Notably, Ser409/410 is at the extreme C-terminal of TDP-43 protein, suggesting a higher level of phosphorylated C-terminal fragments in TREM2 KO mice. c, Whole brain p-hTDP-43 immunoblots of the Sarkosyl-insoluble fraction of WT and TREM2 KO mice at 70 dpi. Western blots were independently repeated twice with n = 4 for each group.

Source data

Extended Data Fig. 4 Phenotypic diversity of microglia in response to TDP-43 pathology by CyTOF.

a, A representative gating strategy illustrating brain myeloid cell being subgated to CD45medCD11b+ microglia. b, Microglia were plotted onto a tSNE. Plots represent distinguishing cell surface markers for microglia of 6 to 8-week-old WT mice. Clustering analysis revealed a major microglia population characterized by CD45mid:CD11b+:CX3CR1hi:F4/80+:CD64+:MERTK+:SiglecH+:CD11c. c, Heat map shows the change of expression levels of typical microglial markers in response to TDP-43 pathology in individual samples. Heat colors of expression levels have been scaled for each marker (blue, low expression; orange, high expression). d, Frequency analysis of microglia based on manual gating of indicated groups at 35 dpi. Significance was calculated using two-way ANOVA, Tukey’s post-hoc analysis. Data represented as mean ± SEM. n.s., not significant, *P < 0.05, **P < 0.01, *** P < 0.001. n = 4 per group, P = 0.0008, F 3,9 = 14.79.

Extended Data Fig. 5 TREM2 deficiency attenuates hTDP-43-induced microglial activation.

hTDP-43 protein expression was induced via intracerebroventricular injection of AAV9.CAG.hTDP-43 in neonatal mouse (AAV9.CAG.Empty as control). a-c, Representative images (a) and quantification of GFP-expressing microglia number (b) and soma size (c) in the primary motor cortex of indicated groups at 35 dpi. M1 and M2 are separated by dashed line. Scale bar, 200 µm. Insets show microglia at higher magnification as indicated by the area in white box. Scale bar, 50 µm. d, Representative images of co-localization of CD11c (white) with Iba1 (red) in microglia phagocytosing GFP-hTDP-43 (green) in the primary motor cortex of WT group at 35 dpi. White arrowheads indicate phagocytic puncta of GFP-hTDP-43. Scale bar, 20 µm. e, Pie chart representing the percentage of microglia phagocytosing GFP-hTDP-43 (green, phagocytic microglia) and those non-phagocytic microglia in d. In b and c, Significance was calculated using two-way ANOVA followed by Tukey’s post hoc test. Data represented as mean ± SEM. n.s., not significant, *P < 0.05, **P < 0.01, *** P < 0.001. b, n = 10 per group, P < 0.0001, F 3,27 = 283.9; c, n = 10 per group, P < 0.0001, F 3,27 = 19.59.

Extended Data Fig. 6 Characterizations of hTDP-43 expression mouse model via local virus injection in the primary motor cortex of adult mice.

GFP-hTDP-43 or hTDP-43 was expressed in the primary motor cortex of 2-month-old mice via stereotactic intracerebral injection of AAV9.CAG.hTDP-43.GFP or AAV9.CAG.hTDP (AAV9.CAG.GFP or AAV9.CAG.Empty as control). a, Schematic picture showing stereotactic virus injection site (upper). Representative image of GFP expression in the primary motor cortex of 2-month-old WT mice at 14 dpi (lower); Dashed lines indicate the borders of layer 4&5. Scale bar, 100 µm. b, Representative images of GFP-hTDP-43 expression in neuronal nuclei (white arrowhead) and diffusely in dendrites (white arrow) in motor cortex of WT mice at 14 dpi. AAV1.CAG.tdTomato virus was co-injected to visualize neurons. Scale bar, 5 µm. c,d, Representative images (c) and quantification (d) of GFP-hTDP-43 expression in the primary motor cortex of WT and TREM2 KO mice at 14 dpi. Scale bar, 100 µm. e, GFP-hTDP-43 immunoblots of primary motor cortex of WT and TREM KO mice at 14 dpi. Western blots were independently repeated four times (n = 8 per group). GAPDH was used as loading control. f,g, Representative images (f) and quantification (g) of GFP-expressing microglia in the primary motor cortex of indicated groups at 14 dpi. Scale bar, 100 µm. h, Representative images of microglia (Iba1, red) phagocytosing GFP-hTDP-43 (green) in the primary motor cortex of indicated groups at 28 dpi. Scale bar, 100 µm. Significance was calculated using either two-tailed unpaired Student’s t-test (d) or two-way ANOVA, Tukey’s post-hoc analysis (g). Data represented as mean ± SEM. n.s., not significant, *P < 0.05, **P < 0.01, *** P < 0.001. d, n = 7 per group, P = 0.4802, t = 0.7286, d.f. = 12; g, n = 12 per group, P < 0.0001, F 3,33 = 133.4.

Source data

Extended Data Fig. 7 TREM2 deficiency facilitates hTDP-43-induced neurodegeneration.

GFP-hTDP-43 or hTDP-43 was expressed in the primary motor cortex of 2-month-old mice via stereotactic intracerebral injection of AAV9.CAG.hTDP-43.GFP or AAV9.CAG.hTDP (AAV9.CAG.GFP or AAV9.CAG.Empty as control). a,b, Representative images (a) and quantification (b) of NeuN immunostaining (red) in the primary motor cortex of indicated groups at 28 dpi. Scale bar, 100 µm. c,d, Representative images (c) and quantification (d) of microglia (GFP) interaction with NeuN+ neurons (red) in the primary motor cortex of indicated groups at 28 dpi. Scale bar, 50 µm. e, Representative images of CD11c (white) expression in microglia (CX3CR1-GFP, green) interacting with NeuN+ neurons (red) in the primary motor cortex of WT groups at 28 dpi. Scale bar, 20 µm. In b and d, Significance was calculated using two-way ANOVA followed by Tukey’s post hoc test. Data represented as mean ± SEM. n.s., not significant, *P < 0.05, **P < 0.01, *** P < 0.001. b, n = 5 per group, P < 0.0001, F 3,12 = 60.56; d, n = 4 per group, P < 0.0001, F 3,9 = 34.58.

Extended Data Fig. 8 TDP-43 can be released from neurons and interact with TREM2 in vitro.

a, Representative images of human iPSC derived neurons infected with AAV9.CAG.hTDP-43.GFP virus or control virus at 21 dpi. AAV1.CAG.tdTomato virus used to visualize neurons. Scale bar, 100 µm. Inserts show neuron morphology at high magnification as indicated by the area in dotted white box. White arrowheads indicate GFP-hTDP-43 translocation in neurites. Scale bar, 20 µm. b, Immunoblots of TDP-43 contained within collected culture media of iPSCs (left) or in pulled down fractions by bead-immobilized GFP antibody (right). c, Representative images of HEK 293 T cells co-transfected with myc-tagged human TREM2 (myc-hTREM2) and GFP-hTDP-43 C terminal fragment (residues 216-414) or GFP control plasmids. Scale bar, 20 µm. d, myc-hTREM2 was co-immunoprecipitated with GFP-hTDP-43 in HEK 293 T cells using bead-immobilized GFP antibody. Experiments were independently repeated three times. e, GFP-hTDP-43 was co-immunoprecipitated with myc-hTREM2 in HEK 293 T cells using bead-immobilized myc antibody. Experiments were independently repeated three times.

Source data

Extended Data Fig. 9 Human TREM2 interacting proteins in HEK293T cells.

a, A schematic illustration of the SILAC methodology to identify human TREM2 interacting proteins in HEK293T cells (Created with BioRender.com). b and c, MS and MS/MS spectra of a peptide ‘D137AGDLWFPGESESFEDAHVEHSISR161’ from human TREM2.

Extended Data Fig. 10 hTDP-43 interacts with TREM2 in vivo in mouse brain.

GFP-hTDP-43 was expressed in the primary motor cortex of 2-month-old mice via stereotactic intracerebral injection of AAV9.CAG.hTDP-43.GFP. a, Representative images of co-localization of TREM2 (red) with Iba1 (white) in microglia phagocytosing GFP-hTDP-43 (green) in the primary motor cortex of indicated groups at 14 dpi. Arrowheads indicate co-localization of TREM2 with phagocytic puncta of GFP-hTDP-43. Scale bar, 20 µm. b, SPR analysis of the binding between recombinant hTREM2 ECD (Met1-Ser174) with NFG and AMM at the indicated concentrations.

Supplementary information

Source data

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xie, M., Liu, Y.U., Zhao, S. et al. TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration. Nat Neurosci 25, 26–38 (2022). https://doi.org/10.1038/s41593-021-00975-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00975-6

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