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:

TREM1 disrupts myeloid bioenergetics and cognitive function in aging and Alzheimer disease mouse models

Abstract

Human genetics implicate defective myeloid responses in the development of late-onset Alzheimer disease. A decline in peripheral and brain myeloid metabolism, triggering maladaptive immune responses, is a feature of aging. The role of TREM1, a pro-inflammatory factor, in neurodegenerative diseases is unclear. Here we show that Trem1 deficiency prevents age-dependent changes in myeloid metabolism, inflammation and hippocampal memory function in mice. Trem1 deficiency rescues age-associated declines in ribose 5-phosphate. In vitro, Trem1-deficient microglia are resistant to amyloid-β42 oligomer-induced bioenergetic changes, suggesting that amyloid-β42 oligomer stimulation disrupts homeostatic microglial metabolism and immune function via TREM1. In the 5XFAD mouse model, Trem1 haploinsufficiency prevents spatial memory loss, preserves homeostatic microglial morphology, and reduces neuritic dystrophy and changes in the disease-associated microglial transcriptomic signature. In aging APPSwe mice, Trem1 deficiency prevents hippocampal memory decline while restoring synaptic mitochondrial function and cerebral glucose uptake. In postmortem Alzheimer disease brain, TREM1 colocalizes with Iba1+ cells around amyloid plaques and its expression is associated with Alzheimer disease clinical and neuropathological severity. Our results suggest that TREM1 promotes cognitive decline in aging and in the context of amyloid pathology.

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: TREM1 deficiency prevents age-associated inflammation and memory decline.
Fig. 2: TREM1 is activated in peripheral MΦ in aging.
Fig. 3: TREM1 suppresses PPP generation of ribose-5P and purine/pyrimidine synthesis in aged MΦ.
Fig. 4: TREM1 deficiency preserves hippocampal function in 5XFAD mice.
Fig. 5: TREM1 deficiency preserves hippocampal spatial memory and brain glucose uptake in APPSwe mice.
Fig. 6: Myeloid TREM1 expression positively associates with increasing AD pathology.

Similar content being viewed by others

Data availability

The data discussed in this publication have been deposited in NCBI’s Gene Expression Omnibus and are accessible through GEO Series accession number GSE229327 for mouse macrophage transcript data and GSE229620 for mouse microglia transcript data. Source data are provided with this paper.

References

  1. Lindenberger, U. Human cognitive aging: corriger la fortune? Science 346, 572–578 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Cunningham, C. & Hennessy, E. Co-morbidity and systemic inflammation as drivers of cognitive decline: new experimental models adopting a broader paradigm in dementia research. Alzheimers Res. Ther. 7, 33 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Franceschi, C. et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev. 128, 92–105 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Nikolich-Zugich, J. The twilight of immunity: emerging concepts in aging of the immune system. Nat. Immunol. 19, 10–19 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Andrews, S. J., Fulton-Howard, B. & Goate, A. Interpretation of risk loci from genome-wide association studies of Alzheimer’s disease. Lancet Neurol. 19, 326–335 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  7. Cruchaga, C. et al. GWAS of cerebrospinal fluid tau levels identifies risk variants for Alzheimer’s disease. Neuron 78, 256–268 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. Melchior, B. et al. Dual induction of TREM2 and tolerance-related transcript, Tmem176b, in amyloid transgenic mice: implications for vaccine-based therapies for Alzheimer’s disease. ASN Neuro 2, e00037 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. 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 

  12. Wang, Y. et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J. Exp. Med. 213, 667–675 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yuan, P. et al. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90, 724–739 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Song, W. M. et al. Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism. J. Exp. Med. 215, 745–760 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Boufenzer, A. et al. TREM-1 mediates inflammatory injury and cardiac remodeling following myocardial infarction. Circ. Res. 116, 1772–1782 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Knapp, S. et al. Cutting edge: expression patterns of surface and soluble triggering receptor expressed on myeloid cells-1 in human endotoxemia. J. Immunol. 173, 7131–7134 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Liu, Q. et al. Peripheral TREM1 responses to brain and intestinal immunogens amplify stroke severity. Nat. Immunol. 20, 1023–1034 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gibot, S. et al. A soluble form of the triggering receptor expressed on myeloid cells-1 modulates the inflammatory response in murine sepsis. J. Exp. Med. 200, 1419–1426 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gibot, S. et al. Plasma level of a triggering receptor expressed on myeloid cells-1: its diagnostic accuracy in patients with suspected sepsis. Ann. Intern. Med. 141, 9–15 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Chaney, A. M. et al. PET imaging of TREM1 identifies CNS-infiltrating myeloid cells in a mouse model of multiple sclerosis. Sci. Transl. Med. 15, eabm6267 (2023).

    Article  CAS  PubMed  Google Scholar 

  22. Colonna, M. TREMs in the immune system and beyond. Nat. Rev. Immunol. 3, 445–453 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Colonna, M. & Facchetti, F. TREM-1 (triggering receptor expressed on myeloid cells): a new player in acute inflammatory responses. J. Infect. Dis. 187, S397–S401 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Bouchon, A., Dietrich, J. & Colonna, M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J. Immunol. 164, 4991–4995 (2000).

    Article  CAS  PubMed  Google Scholar 

  25. Netea, M. G. et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) amplifies the signals induced by the NACHT-LRR (NLR) pattern recognition receptors. J. Leukoc. Biol. 80, 1454–1461 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Tammaro, A. et al. TREM-1 and its potential ligands in non-infectious diseases: from biology to clinical perspectives. Pharmacol. Ther. 177, 81–95 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Replogle, J. M. et al. A TREM1 variant alters the accumulation of Alzheimer-related amyloid pathology. Ann. Neurol. 77, 469–477 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu, Y. S. et al. Common variant in TREM1 influencing brain amyloid deposition in mild cognitive impairment and Alzheimer’s disease. Neurotox. Res. 37, 661–668 (2020).

    Article  CAS  PubMed  Google Scholar 

  29. Minhas, P. S. et al. Restoring metabolism of myeloid cells reverses cognitive decline in ageing. Nature 590, 122–128 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. Flowers, A., Bell-Temin, H., Jalloh, A., Stevens, S. M. & Bickford, P. C. Proteomic analysis of aged microglia: shifts intranscription, bioenergetics, and nutrient response. J. Neuroinflammation 14, 96 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Rath, S. et al. MitoCarta3.0: an updated mitochondrial proteome now with sub-organelle localization and pathway annotations. Nucleic Acids Res. 49, D1541–D1547 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Fox, D. B. et al. NRF2 activation promotes the recurrence of dormant tumour cells through regulation of redox and nucleotide metabolism. Nat. Metab. 2, 318–334 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang, H., Davies, K. J. A. & Forman, H. J. Oxidative stress response and Nrf2 signaling in aging. Free Radic. Biol. Med. 88, 314–336 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lee, W. J. et al. Regional Aβ-tau interactions promote onset and acceleration of Alzheimer’s disease tau spreading. Neuron 110, 1932–1943.e5 (2022).

  36. Oblak, A. L. et al. Comprehensive evaluation of the 5XFAD mouse model for preclinical testing applications: a MODEL-AD study. Front. Aging Neurosci. 13, 713726 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Borchelt, D. R. et al. Accelerated amyloid deposition in the brains of transgenic mice co-expressing mutant presenilin 1 and amyloid precursor protein. Neuron 19, 939–945 (1997).

    Article  CAS  PubMed  Google Scholar 

  38. Jain, N., Lewis, C. A., Ulrich, J. D. & Holtzman, D. M. Chronic TREM2 activation exacerbates Aβ-associated tau seeding and spreading. J. Exp. Med. 220, e20220654 (2023).

    Article  CAS  PubMed  Google Scholar 

  39. Hsiao, K. et al. Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice. Science 274, 99–102 (1996).

    Article  CAS  PubMed  Google Scholar 

  40. Gauba, E., Chen, H., Guo, L. & Du, H. Cyclophilin D deficiency attenuates mitochondrial F1Fo ATP synthase dysfunction via OSCP in Alzheimer’s disease. Neurobiol. Dis. 121, 138–147 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Bouter, C. & Bouter, Y. 18F-FDG-PET in mouse models of Alzheimer’s disease. Front. Med. (Lausanne) 6, 71 (2019).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  43. Rajkowska, G. & Goldman-Rakic, P. S. Cytoarchitectonic definition of prefrontal areas in the normal human cortex: II. Variability in locations of areas 9 and 46 and relationship to the Talairach Coordinate System. Cereb. Cortex 5, 323–337 (1995).

    Article  CAS  PubMed  Google Scholar 

  44. Thompson, P. M. et al. Cortical change in Alzheimer’s disease detected with a disease-specific population-based brain atlas. Cereb. Cortex 11, 1–16 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Clinton, J., Blackman, S. E., Royston, M. C. & Roberts, G. W. Differential synaptic loss in the cortex in Alzheimer’s disease: a study using archival material. Neuroreport 5, 497–500 (1994).

    Article  CAS  PubMed  Google Scholar 

  46. Edler, M. K., Mhatre-Winters, I. & Richardson, J. R. Microglia in aging and Alzheimer’s disease: a comparative species review. Cells 10, 1138 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wilson, E. N. et al. Soluble TREM2 is elevated in Parkinson’s disease subgroups with elevated CSF tau. Brain 143, 932–943 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Suarez-Calvet, M. et al. sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early-stage Alzheimer’s disease and associate with neuronal injury markers. EMBO Mol. Med. 8, 466–476 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Shi, X., Wei, T., Hu, Y., Wang, M. & Tang, Y. The associations between plasma soluble Trem1 and neurological diseases: a Mendelian randomization study. J. Neuroinflammation 19, 218 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ferkingstad, E. et al. Large-scale integration of the plasma proteome with genetics and disease. Nat. Genet. 53, 1712–1721 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Schwartzentruber, J. et al. Genome-wide meta-analysis, fine-mapping and integrative prioritization implicate new Alzheimer’s disease risk genes. Nat. Genet. 53, 392–402 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Xiang, X. et al. Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases. Sci. Transl. Med. 13, eabe5640 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Zimmer, E. R., Pascoal, T. A., Rosa-Neto, P., Nordberg, A. & Pellerin, L. Comment on ‘Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases’. Sci. Transl. Med. 14, eabm8302 (2022).

    Article  CAS  PubMed  Google Scholar 

  54. Weber, B. et al. TREM-1 deficiency can attenuate disease severity without affecting pathogen clearance. PLoS Pathog. 10, e1003900 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Yousef, H. et al. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat. Med. 25, 988–1000 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang, L. et al. Peak annotation and verification engine for untargeted LC-MS metabolomics. Anal. Chem. 91, 1838–1846 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Adusumilli, R. & Mallick, P. Data conversion with ProteoWizard msConvert. Methods Mol. Biol. 1550, 339–368 (2017).

    Article  CAS  PubMed  Google Scholar 

  59. Clasquin, M. F., Melamud, E. & Rabinowitz, J. D. LC-MS data processing with MAVEN: a metabolomic analysis and visualization engine. Curr. Protoc. Bioinformatics Chapter 14, Unit14 11 (2012).

    PubMed  Google Scholar 

  60. Melamud, E., Vastag, L. & Rabinowitz, J. D. Metabolomic analysis and visualization engine for LC-MS data. Anal. Chem. 82, 9818–9826 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pang, Z. et al. MetaboAnalyst 5.0: narrowing the gap between raw spectra and functional insights. Nucleic Acids Res. 49, W388–W396 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Young, K. & Morrison, H. Quantifying microglia morphology from photomicrographs of immunohistochemistry prepared tissue using ImageJ. J. Vis. Exp. 136, e57648 (2018).

    Google Scholar 

  63. Chaney, A. M., Johnson, E. M., Cropper, H. C. & James, M. L. PET imaging of neuroinflammation using [11C]DPA-713 in a mouse model of ischemic stroke. J. Vis. Exp. https://doi.org/10.3791/57243 (2018).

  64. Casali, B. T. & Landreth, G. E. Aβ extraction from murine brain homogenates. Bio Protoc. 6, e1787 (2016).

  65. Beach, T. G. et al. Arizona Study of Aging and Neurodegenerative Disorders and Brain and Body Donation Program. Neuropathology 35, 354–389 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. The National Institute on Aging, and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease. Neurobiol. Aging 18, S1–S2 (1997).

  67. McKeith, I. G. et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology 65, 1863–1872 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Dickson, D. W. Required techniques and useful molecular markers in the neuropathologic diagnosis of neurodegenerative diseases. Acta Neuropathol. 109, 14–24 (2005).

    Article  PubMed  Google Scholar 

  69. Hemani, G., Tilling, K. & Davey Smith, G. Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 13, e1007081 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Hemani, G. et al. The MR-Base platform supports systematic causal inference across the human phenome. eLife 7, e34408 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Liu, Q. et al. PGE2 signaling via the neuronal EP2 receptor increases injury in a model of cerebral ischemia. Proc. Natl Acad. Sci. USA 116, 10019–10024 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Iweka, C. A. et al. Myeloid deficiency of the intrinsic clock protein BMAL1 accelerates cognitive aging by disrupting microglial synaptic pruning. J. Neuroinflammation 20, 48 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Blacher, E. et al. Aging disrupts circadian gene regulation and function in macrophages. Nat. Immunol. 23, 229–236 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grant no. RF1AG053001K (K.I.A.), grant no. RO1NS100180 (K.I.A.), grant no. RF1AG070131 (K.I.A.), grant no. RF1AG070839 (K.I.A.), grant no. P30 AG0066515 (K.I.A.), American Heart Foundation grant no. 19PABH1345800 (K.I.A.), the Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University (K.I.A.), The Archer Foundation (K.I.A.), Stanford School of Medicine Dean’s Postdoctoral Fellowship (E.N.W. and E.B.), HHMI Hanna H. Gray Fellows Program (M.R.M.), Burroughs Wellcome Fund PDEP (M.R.M.), an Alzheimer’s Association Research Fellowship (K.A.Z.), the Paul and Daisy Soros Fellowship for New Americans (P.S.M.), the Gerald J. Lieberman Fellowship (P.S.M.) and Marie Skłodowska-Curie Grant no. 888494 (E.B.). K.I.A. is a Chan Zuckerberg–San Francisco Biohub Investigator. Data and tissue were obtained from the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND) (https://www.bannerhealth.com/services/research/about-banner-research/research-programs/brain-and-body-donation-program) supported by grant no. U24NS072026, grant no. P30AG19610, the Arizona Department of Health Services, the Arizona Biomedical Research Commission and the Michael J. Fox Foundation for Parkinson’s Research. We thank the Stanford Cyclotron & Radiochemistry Facility, the Stanford SCi3 Small-Animal Imaging Facility, the Stanford Shared FACS Facility, Stanford Human Immune Monitoring Core and the Stanford Neuroscience Microscopy Service.

Author information

Authors and Affiliations

Authors

Contributions

E.N.W. and K.I.A. conceived and planned this study. E.N.W., T.G.B., M.D.G., M.L.J., M.S.B., J.D.R. and K.I.A. contributed to supervising experimental design. E.N.W., C.W., H.E.E., M.S.S., M.R.M., Q.W., K.A.Z., A.C., J.A.R.B., E.G., P.S.M., E.B., Y.J.T., C.A.I., Y.L.G., M.P., H.C., P.J., Q.L., S.S.M., A.J.Z., M.X., J.U., J.H., A.S.D. and G.E.S. conducted the experiments. E.N.W., C.W., H.E.E., M.S.S., A.C., J.A.R.B., E.G., P.S.M., Y.L.G., A.J.Z., M.X., J.U. and J.H. performed collection of the data and statistical analysis. E.N.W. and K.I.A. wrote the manuscript. All authors reviewed and approved of the manuscript.

Corresponding author

Correspondence to Katrin I. Andreasson.

Ethics declarations

Competing interests

M.L.J. and K.I.A. are co-founders and Scientific Advisory Board members of Willow Neuroscience, Inc. The other authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Michael Heneka, Inhee Mook-Jung and the other, anonymous, reviewer(s) 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 Flow cytometry gating for quantification of TREM1 expression in blood, spleen and brain myeloid cells.

a Gating strategy for live CD45 + Cd11b + TREM1+ blood myeloid cells. b Gating strategy for live CD45 + Cd11b + TREM1+ spleen myeloid cells. c Gating strategy for live CD45loCd11b + TREM1+ microglia; also includes gating for P2RY12, CD206, CX3CR1 and Tmem119 microglial markers.

Extended Data Fig. 2 TREM1 amplifies levels of inflammatory factors but does not alter phagocytosis.

a Quantification of immune factors in lysates of peritoneal MΦ isolated from 8.5 mo WT and Trem1-/- mice treated with vehicle or 100 ng/ml LPS for 20 hr. Two-way ANOVA with Tukey’s multiple comparisons shown for WT + LPS vs Trem1-/-+ LPS (n = 5–6 male mice per genotype as shown). b Trem1 deficiency does not alter E.coli phagocytosis in peritoneal MΦ from 2–3 mo male mice. Cells were stimulated with vehicle or increasing concentrations of LPS for 20 h. 2-way ANOVA, effect of LPS ***P < 0.001, no effect of Trem1 genotype basally or with LPS stimulation on 6–11 technical replicate wells as shown.

Source data

Extended Data Fig. 3 TREM1 promotes a pro-inflammatory polarization state in aged peritoneal MΦ.

a Gating strategy for live Cd11b+Ly6GCD45+Ly6CHi and Cd11b+Ly6GCD45+Ly6CLo peritoneal MΦ from young WT and Trem1-/- (2 mo) and aged WT and Trem1-/- (22–23 mo) male mice. b Representative gating for MHC II, CD86, and CD71 positive Ly6CHi peritoneal MΦ. c Representative gating for MHC II, CD86, and CD71 positive Ly6CLo peritoneal MΦ. d Multianalyte Luminex quantification of immune factors in young WT (2 mo) and aged WT (23–24 mo) and aged Trem1-/- (23–24 mo) cerebral cortex. One-way ANOVA with Tukey’s multiple comparisons shown (n = 5–6 male mice per group as shown). e Total time exploring (in seconds) during the training phase of the NOR task; ANOVA with Tukey’s multiple comparisons show no significant differences (n = 6–8 female mice as shown). f Speed (pixels/second) of mice during the final trial of the Barnes Maze; ANOVA with Tukey’s multiple comparisons (n = 5–6 male and female mice as shown).

Source data

Extended Data Fig. 4 Trem1 deficiency alters gene expression in aged peripheral MΦ with minimal changes in aged brain microglia.

a Venn diagram of DEGs (all genes q < 0.05) in pairwise comparisons of microglia isolated from aged (18 - 20 mo) vs young (3 mo) mice and pairwise comparisons of microglia isolated from aged Trem1-/- vs aged WT mice (18–20 mo). Blue indicates number of downregulated genes while red indicates the number of upregulated genes (n = 3 microglial samples (2 pooled male mice per sample) per group). b KEGG Pathway enrichment analysis of upregulated genes in aged compared to young microglia; there were no enriched pathways in the comparison of aged WT vs aged Trem1-/- microglia. c Venn diagrams showing the number of DEGs (all genes q < 0.05) in pairwise comparisons of primary mouse MΦ from young WT (2 mo), aged WT (25 mo) and aged Trem1-/- (25 mo) mice. Blue indicates number of downregulated genes while red indicates the number of upregulated genes (n = 3–4 male mice per group). d Pathway enrichment analysis of upregulated genes in aged MΦ vs young and in aged Trem1-/- vs aged WT MΦ. e Heatmap of DEGs (q < 0.05) comprising the Coordinated Lysosomal Expression and Regulation (CLEAR) network. Lysosomal functional categories are indicated for each gene. Aged Trem1-/- mice cluster with the young WT mice. Scale represents z-score values from FPKM. n = 3–4 male mice per group.

Source data

Extended Data Fig. 5 TREM1 deficiency restores youthful metabolism in aged macrophages.

a PCA of metabolites from young (2 mo) WT and Trem1-/- peritoneal MΦ (n = 3 male mice per group). b PCA of significantly regulated metabolites from primary peritoneal MΦ isolated from young WT (2 mo), aged WT (25 mo) and aged Trem1-/- (25 mo) male mice. n = 3–5 male mice per group. c Volcano plot comparing aged WT vs young WT (left) and aged Trem1-/- vs aged WT (right) peritoneal MΦ with Log2 fold change (FC) and -Log10(P) values of metabolites. Significantly regulated metabolites with -log10(P) > 2 and log2 fold change > 1 are shown in blue. d KEGG metabolic pathway enrichment analysis of differentially regulated metabolites between aged WT and young WT macrophages and between aged Trem1-/- vs aged WT MΦ. X-axis shows -Log10(q) value. No pathways were significant following FDR correction for the comparison of aged Trem1-/- vs young WT. Pathway analysis was performed using MetaboAnalyst 5.0; n = 3–5 male mice per group. e Total ion count of Ribose-5P, the precursor of purines and pyrimidines and total ion counts for nucleobases and derivatives, ribo-nucleosides, deoxynucleosides, and nucleoside monophosphates. Data are analyzed by one-way ANOVA with Tukey’s multiple comparisons (n = 3–5 male mice per group). Abbreviations: AMP: adenosine monophosphate, GMP: guanosine monophosphate, UMP: uridine monophosphate, CMP cytidine monophosphate, IMP: inosine monophosphate. n = 3–5 male mice per group as shown. f Transcription factor (TF) enrichment analysis revealing TFs enriched for differentially expressed metabolite enzymes. g FKPM of NRF2. ANOVA followed by Tukey’s multiple comparison (n = 3–4 male mice per group as shown). h FKPM of the 3 pyruvate dehydrogenase complex (PDH) subunits: pyruvate dehydrogenase (PDHA1), dihydrolipoyl transacetylase (DLAT) and dihydrolipoyl dehydrogenase (DLD); ANOVA followed by Tukey’s multiple comparison (n = 3–4 male mice per group as shown).

Source data

Extended Data Fig. 6 TREM1 expression does not significantly change in context of accumulating amyloid.

a (Left) Immunofluorescent staining of TREM1 in 9 mo WT and APPSwe-PS1∆E9 hippocampal stratum lacunosum in IBA1+microglia. 5–7 hippocampal sections per mouse were imaged, n = 5 male mice/genotype. Scale bar = 10 µm. TREM1 colocalizes with Iba1+ cells (white arrows). (Right) Quantification of MFI. b Thioflavin-S (ThioS) fluorescent staining of hippocampus in 9–10 mo 5XFAD and 5XFAD;Trem1+/- female mice. Scale bar = 500 µm. c Soluble levels of cerebral cortical Aß40 and 42 and the ratio of Aß42/40 in 5X FAD and 5XFAD;Trem1 + /- mice. Student’s two tailed t-test (n = 4–6 males/group as shown, 10–13 mo). d Insoluble levels of cerebral cortical Aß40 and 42 and the ratio of Aß42/40 in 5X FAD and 5XFAD;Trem1 + /- mice; Student’s two tailed t-test (n = 4–6 males/group as shown, 10–13 mo). e Quantification of hippocampal immune factors in WT, 5XFAD, and 5XFAD;Trem1+/- 10 mo mice. One-way ANOVA with Tukey post-hoc comparisons (n = 5–6 female mice per group as shown). f Microglial number from Fig. 4i. One-way ANOVA with Tukey’s multiple comparisons (n = 5 male mice per condition).

Source data

Extended Data Fig. 7 Disease-associated microglial (DAM) signature in 5XFAD;Trem1+/- mice.

a PCA of significantly regulated DAM signature genes from primary microglia isolated from WT, 5XFAD and 5XFAD/Trem1+/- mice. Individual microglial samples were pooled from 2 male mice (n = 3 samples per condition). b Hierarchical clustering of DAM signature gene set showing homeostatic, DAM Stage 1 and DAM Stage 2 genes. Scale represents z-score values from FPKM. c Hierarchical clustering of top 170 DEGs (FDR-corrected) belonging to the full DAM signature gene set. Scale represents z-score values from FPKM. Scale represents z-score values from FPKM. d (Left) Percent of CX3CR1+ microglia in young (3 mo) and 13–17 mo WT, 5xFAD and 5xFAD;Trem1+/- mice. (Right) MFI of CX3CR1+ microglia. One-way ANOVA with Tukey’s multiple comparison, not significant (n = 5–8 male and female mice per group as shown). e (Left) Percent of Tmem119+ microglia in young (3 mo) and 13–17 mo WT, 5xFAD and 5xFAD;Trem1+/- mice. (Right) MFI of Tmem119+ microglia. One-way ANOVA with Tukey’s multiple comparisons (n = 4-5 mice per group). f (Left) Total time exploring (in seconds) during the training phase of the NOR task in APPSwe mice; ANOVA with Tukey’s post-hoc test (n = 14-17 male and female mice per condition as shown). g Motor speed (pixels/sec) during the final training session of the Barnes maze; ANOVA with Tukey’s post-hoc test (n = 6 male and female mice per group). h Distance traveled during the final training session of the Barnes maze; ANOVA with Tukey’s post-hoc test (n = 6 male and female mice per group). i Coupling assay tracings of synaptic mitochondria oxygen consumption rates (OCR). Shown over time are the rates of basal Complex II respiration, State III (ADP stimulated respiration), State IV (oligomycin) and State IIIu (State III uncoupled, FCCP) that were consecutively measured over the course of the assay (n = 5-9 male mice per condition). j Diagram of model of action of TREM1 in aging and transgenic mice with amyloid accumulation. In aging, peripheral TREM1 activity contributes to declines in myeloid metabolism and immune functions that lead to age-dependent cognitive decline. In models of amyloid accumulation, both peripheral and microglial TREM1 contribute to cognitive deficits associated with local accumulation of amyloid.

Source data

Extended Data Fig. 8 TREM1 expression in development of AD.

a Immunofluorescent staining of human frontal cortex without addition of TREM1 antibody and secondary only control (red), Iba1 (green), and X-34 (blue) signal. This antibody validation experiment was performed three times. Scale bar = 20 μm. b Immunoblot of TREM1 protein in postmortem human mid frontal gyrus. Clinicopathological diagnoses: non-demented Braak I-II, demented non-AD Braak stages I-II, AD Braak III-IV and AD Braak V-VI. Primary antibody detected human TREM1 band at the molecular weight of positive control (human liver lysates, Cat# HT-314, Zyagen, San Diego, CA). The TREM1 band used for analysis are indicated by blue box. Also shown is the band for ß-actin at 42 kDa. This antibody validation experiment was performed once. c Immunoblot of TREM2 protein in postmortem human mid frontal gyrus. Primary antibody detects human TREM2 band at the molecular weight of positive control (human liver lysates, Cat# HT-314, Zyagen, San Diego, CA). TREM2 bands used for analysis are indicated by blue box. Also shown is the band for ß-actin at 42 kDa. This antibody validation experiment was performed once. d Mendelian Randomization (MR) analyses with Alzheimer’s disease (AD) risk level as exposure and sTREM1 and sTREM2 plasma protein level as outcomes. Blue lines are estimated MR-median weight effects, and the dashed lines indicate the 95% confidence interval for the MR effects.

Source data

Extended Data Table 1 List of Metabolites Detected by LC-MS
Extended Data Table 2 Demographic Characteristics of Study Participants

Supplementary information

Source data

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wilson, E.N., Wang, C., Swarovski, M.S. et al. TREM1 disrupts myeloid bioenergetics and cognitive function in aging and Alzheimer disease mouse models. Nat Neurosci (2024). https://doi.org/10.1038/s41593-024-01610-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41593-024-01610-w

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