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:

Alzheimer’s disease modification mediated by bone marrow-derived macrophages via a TREM2-independent pathway in mouse model of amyloidosis

Abstract

Microglia and monocyte-derived macrophages (MDM) are key players in dealing with Alzheimer’s disease. In amyloidosis mouse models, activation of microglia was found to be TREM2 dependent. Here, using Trem2−/−5xFAD mice, we assessed whether MDM act via a TREM2-dependent pathway. We adopted a treatment protocol targeting the programmed cell death ligand-1 (PD-L1) immune checkpoint, previously shown to modify Alzheimer’s disease via MDM involvement. Blockade of PD-L1 in Trem2−/−5xFAD mice resulted in cognitive improvement and reduced levels of water-soluble amyloid beta1–42 with no effect on amyloid plaque burden. Single-cell RNA sequencing revealed that MDM, derived from both Trem2−/− and Trem2+/+5xFAD mouse brains, express a unique set of genes encoding scavenger receptors (for example, Mrc1, Msr1). Blockade of monocyte trafficking using anti-CCR2 antibody completely abrogated the cognitive improvement induced by anti-PD-L1 treatment in Trem2−/−5xFAD mice and similarly, but to a lesser extent, in Trem2+/+5xFAD mice. These results highlight a TREM2-independent, disease-modifying activity of MDM in an amyloidosis mouse model.

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: DAM are derived from resident microglia and their level is elevated following anti-PD-L1 treatment.
Fig. 2: Treatment with anti-PD-L1 antibody reduces cognitive deficits in a Trem2-independent manner.
Fig. 3: Treatment with anti-PD-L1 antibody reduces the levels of TBS-soluble Aβ1–42 in a Trem2-independent manner.
Fig. 4: MDM share a unique transcriptomic signature in both Trem2+/+5xFAD and Trem2–/–5xFAD brain.
Fig. 5: Elimination of monocytes using CCR2-blocking antibody abrogates the beneficial effects of anti-PD-L1 treatment on both cognition and TBS-soluble Aβ1–42.
Fig. 6: Blockade of CCR2 completely abrogates the beneficial effect of anti-PD-L1 treatment assessed by NOR, but only partially when assessed by RAWM, in Trem2+/+5xFAD mice.

Similar content being viewed by others

Data availability

The single-cell RNA-seq data were deposited at the National Center for Biotechnology Information’s GEO with accession no. GSE176085. For the differential gene expression analysis presented in Extended Data Fig. 1b, we used the single-cell RNA-seq dataset published by Keren-Shaul et al.20 available at GEO with accession no. GSE176085. All underlying data used for generation of figures are collated in the associated source files. All other data are available from the corresponding authors upon reasonable request.

Code availability

Metacell source code can be found at https://github.com/tanaylab/metacell. Source code used for single-cell RNA-seq analysis can be found at https://bitbucket.org/amitlab/AD_aPDL1_TREM2.

References

  1. Masters, C. L. et al. Alzheimer’s disease. Nat. Rev. Dis. Primers 1, 15056 (2015).

    Article  PubMed  Google Scholar 

  2. Masters, C. L. & Selkoe, D. J. Biochemistry of amyloid beta-protein and amyloid deposits in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2, a006262 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Lesne, S. et al. A specific amyloid-beta protein assembly in the brain impairs memory. Nature 440, 352–357 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Bernstein, S. L. et al. Amyloid-beta protein oligomerization and the importance of tetramers and dodecamers in the aetiology of Alzheimer’s disease. Nat. Chem. 1, 326–331 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Shea, D. et al. alpha-Sheet secondary structure in amyloid beta-peptide drives aggregation and toxicity in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 116, 8895–8900 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sengupta, U., Nilson, A. N. & Kayed, R. The role of amyloid-beta oligomers in toxicity, propagation, and immunotherapy. EBioMed. 6, 42–49 (2016).

    Article  Google Scholar 

  7. Selkoe, D. J. & Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ferreira, S. T., Lourenco, M. V., Oliveira, M. M., De & Felice, F. G. Soluble amyloid-beta oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front. Cell. Neurosci. 9, 191 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Panza, F., Lozupone, M., Logroscino, G. & Imbimbo, B. P. A critical appraisal of amyloid-beta-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 15, 73–88 (2019).

    Article  PubMed  Google Scholar 

  10. Giacobini, E. & Gold, G. Alzheimer disease therapy–moving from amyloid-beta to tau. Nat. Rev. Neurol. 9, 677–686 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Congdon, E. E. & Sigurdsson, E. M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 14, 399–415 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Simard, A. R., Soulet, D., Gowing, G., Julien, J. P. & Rivest, S. Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron 49, 489–502 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Town, T. et al. Blocking TGF-beta-Smad2/3 innate immune signaling mitigates Alzheimer-like pathology. Nat. Med. 14, 681–687 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dong, Y., Li, X., Cheng, J. & Hou, L. Drug development for Alzheimer’s disease: microglia induced neuroinflammation as a target? Int. J. Mol. Sci. 20, 558 (2019).

  16. Butovsky, O. et al. Glatiramer acetate fights against Alzheimer’s disease by inducing dendritic-like microglia expressing insulin-like growth factor 1. Proc. Natl Acad. Sci. USA 103, 11784–11789 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. Baruch, K. et al. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease. Nat. Med. 22, 135–137 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Rosenzweig, N. et al. PD-1/PD-L1 checkpoint blockade harnesses monocyte-derived macrophages to combat cognitive impairment in a tauopathy mouse model. Nat. Commun. 10, 465 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. Da Mesquita, S. et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 560, 185–191 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Koronyo, Y. et al. Therapeutic effects of glatiramer acetate and grafted CD115+ monocytes in a mouse model of Alzheimer’s disease. Brain 138, 2399–2422 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Wynn, T. A. & Vannella, K. M. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 44, 450–462 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Minutti, C. M., Knipper, J. A., Allen, J. E. & Zaiss, D. M. Tissue-specific contribution of macrophages to wound healing. Semin. Cell Dev. Biol. 61, 3–11 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Peiser, L., Mukhopadhyay, S. & Gordon, S. Scavenger receptors in innate immunity. Curr. Opin. Immunol. 14, 123–128 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Ginhoux, F. & Prinz, M. Origin of microglia: current concepts and past controversies. Cold Spring Harb. Perspect. Biol. 7, a020537 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Hanisch, U. K. & Kettenmann, H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 10, 1387–1394 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Oakley, H. et al. Intraneuronal beta-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 

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

  30. Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W. & Rossi, F. M. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 10, 1538–1543 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Shechter, R. et al. Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med. 6, e1000113 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Shechter, R. et al. Recruitment of beneficial M2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus. Immunity 38, 555–569 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gadani, S. P., Walsh, J. T., Smirnov, I., Zheng, J. & Kipnis, J. The glia-derived alarmin IL-33 orchestrates the immune response and promotes recovery following CNS injury. Neuron 85, 703–709 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Koronyo-Hamaoui, M. et al. Attenuation of AD-like neuropathology by harnessing peripheral immune cells: local elevation of IL-10 and MMP-9. J. Neurochem. 111, 1409–1424 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Naert, G. & Rivest, S. Hematopoietic CC-chemokine receptor 2 (CCR2) competent cells are protective for the cognitive impairments and amyloid pathology in a transgenic mouse model of Alzheimer’s disease. Mol. Med. 18, 297–313 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Ben-Yehuda, H. et al. Key role of the CCR2-CCL2 axis in disease modification in a mouse model of tauopathy. Mol. Neurodegener. 16, 39 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Xing, Z. et al. Influenza vaccine combined with moderate-dose PD1 blockade reduces amyloid-beta accumulation and improves cognition in APP/PS1 mice. Brain Behav. Immun. 91, 128–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Zuroff, L., Daley, D., Black, K. L. & Koronyo-Hamaoui, M. Clearance of cerebral Abeta in Alzheimer’s disease: reassessing the role of microglia and monocytes. Cell. Mol. Life Sci. 74, 2167–2201 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dionisio-Santos, D. A., Olschowka, J. A. & O’Banion, M. K. Exploiting microglial and peripheral immune cell crosstalk to treat Alzheimer’s disease. J. Neuroinflammation 16, 74 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Li, Q. & Barres, B. A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 18, 225–242 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Yu, C., Roubeix, C., Sennlaub, F. & Saban, D. R. Microglia versus monocytes: distinct roles in degenerative diseases of the retina. Trends Neurosci. 43, 433–449 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Koronyo-Hamaoui, M. et al. Peripherally derived angiotensin converting enzyme-enhanced macrophages alleviate Alzheimer-related disease. Brain 143, 336–358 (2020).

    Article  PubMed  Google Scholar 

  43. Frenkel, D. et al. Scara1 deficiency impairs clearance of soluble amyloid-beta by mononuclear phagocytes and accelerates Alzheimer’s-like disease progression. Nat. Commun. 4, 2030 (2013).

    Article  PubMed  Google Scholar 

  44. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Keren-Shaul, H. et al. MARS-seq2.0: an experimental and analytical pipeline for indexed sorting combined with single-cell RNA sequencing. Nat. Protoc. 14, 1841–1862 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Baran, Y. et al. MetaCell: analysis of single-cell RNA-seq data using K-nn graph partitions. Genome Biol. 20, 206 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Griciuc, A. et al. TREM2 acts downstream of CD33 in modulating microglial pathology in Alzheimer’s disease. Neuron 103, 820–835 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Meilandt, W. J. et al. Trem2 deletion reduces late-stage amyloid plaque accumulation, elevates the Abeta42:Abeta40 ratio, and exacerbates axonal dystrophy and dendritic spine loss in the PS2APP Alzheimer’s mouse model. J. Neurosci. 40, 1956–1974 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, J., Tanila, H., Puolivali, J., Kadish, I. & van Groen, T. Gender differences in the amount and deposition of amyloidbeta in APPswe and PS1 double transgenic mice. Neurobiol. Dis. 14, 318–327 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Bundy, J. L., Vied, C., Badger, C. & Nowakowski, R. S. Sex-biased hippocampal pathology in the 5XFAD mouse model of Alzheimer’s disease: a multi-omic analysis. J. Comp. Neurol. 527, 462–475 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Carroll, J. C. et al. Sex differences in beta-amyloid accumulation in 3xTg-AD mice: role of neonatal sex steroid hormone exposure. Brain Res. 1366, 233–245 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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  PubMed  PubMed Central  Google Scholar 

  54. Benilova, I., Karran, E., De & Strooper, B. The toxic Abeta oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat. Neurosci. 15, 349–357 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Shichita, T. et al. MAFB prevents excess inflammation after ischemic stroke by accelerating clearance of damage signals through MSR1. Nat. Med. 23, 723–732 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. 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-beta by microglia. Neuron 91, 328–340 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. London, A. et al. Neuroprotection and progenitor cell renewal in the injured adult murine retina requires healing monocyte-derived macrophages. J. Exp. Med. 208, 23–39 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

  59. Jordao, M. J. C. et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 363, eaat7554 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Zhou, Y. et al. Mannose receptor modulates macrophage polarization and allergic inflammation through miR-511-3p. J. Allergy Clin. Immunol. 141, 350–364 (2018).

    Article  CAS  PubMed  Google Scholar 

  61. Xu, Z. J. et al. The M2 macrophage marker CD206: a novel prognostic indicator for acute myeloid leukemia. Oncoimmunology 9, 1683347 (2020).

    Article  PubMed  Google Scholar 

  62. Subramanian, K. et al. Pneumolysin binds to the mannose receptor C type 1 (MRC-1) leading to anti-inflammatory responses and enhanced pneumococcal survival. Nat. Microbiol. 4, 62–70 (2019).

    Article  CAS  PubMed  Google Scholar 

  63. Dani, N. et al. A cellular and spatial map of the choroid plexus across brain ventricles and ages. Cell 184, 3056–3074 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. 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 

  65. Reed-Geaghan, E. G., Croxford, A. L., Becher, B. & Landreth, G. E. Plaque-associated myeloid cells derive from resident microglia in an Alzheimer’s disease model. J. Exp. Med. 217, e20191374 (2020).

  66. Schoch, K. M. et al. Acute Trem2 reduction triggers increased microglial phagocytosis, slowing amyloid deposition in mice. Proc. Natl Acad. Sci. USA 118, e2100356118 (2021).

  67. Hu, Y. et al. Replicative senescence dictates the emergence of disease-associated microglia and contributes to Abeta pathology. Cell Rep. 35, 109228 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. McDonald, J. M., Cairns, N. J., Taylor-Reinwald, L., Holtzman, D. & Walsh, D. M. The levels of water-soluble and triton-soluble Abeta are increased in Alzheimer’s disease brain. Brain Res. 1450, 138–147 (2012).

    Article  CAS  PubMed  Google Scholar 

  69. Latta-Mahieu, M. et al. Systemic immune-checkpoint blockade with anti-PD1 antibodies does not alter cerebral amyloid-beta burden in several amyloid transgenic mouse models. Glia 66, 492–504 (2018).

    Article  PubMed  Google Scholar 

  70. Nelson, P. T. et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J. Neuropathol. Exp. Neurol. 71, 362–381 (2012).

    Article  PubMed  Google Scholar 

  71. Tao, Q. et al. Association of chronic low-grade inflammation with risk of Alzheimer disease in ApoE4 carriers. JAMA Netw. Open 1, e183597 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Misiak, B., Leszek, J. & Kiejna, A. Metabolic syndrome, mild cognitive impairment and Alzheimer’s disease–the emerging role of systemic low-grade inflammation and adiposity. Brain Res. Bull. 89, 144–149 (2012).

    Article  CAS  PubMed  Google Scholar 

  73. Ulland, T. K. et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell 170, 649–663 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mall, C. et al. Repeated PD-1/PD-L1 monoclonal antibody administration induces fatal xenogeneic hypersensitivity reactions in a murine model of breast cancer. Oncoimmunology 5, e1075114 (2016).

    Article  PubMed  Google Scholar 

  75. Mack, M. et al. Expression and characterization of the chemokine receptors CCR2 and CCR5 in mice. J. Immunol. 166, 4697–4704 (2001).

    Article  CAS  PubMed  Google Scholar 

  76. Bruhl, H. et al. Targeting of Gr-1+,CCR2+ monocytes in collagen-induced arthritis. Arthritis Rheum. 56, 2975–2985 (2007).

    Article  PubMed  Google Scholar 

  77. Baruch, K. et al. Breaking immune tolerance by targeting Foxp3(+) regulatory T cells mitigates Alzheimer’s disease pathology. Nat. Commun. 6, 7967 (2015).

    Article  CAS  PubMed  Google Scholar 

  78. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  79. Alamed, J., Wilcock, D. M., Diamond, D. M., Gordon, M. N. & Morgan, D. Two-day radial-arm water maze learning and memory task; robust resolution of amyloid-related memory deficits in transgenic mice. Nat. Protoc. 1, 1671–1679 (2006).

    Article  CAS  PubMed  Google Scholar 

  80. Bevins, R. A. & Besheer, J. Object recognition in rats and mice: a one-trial non-matching-to-sample learning task to study ‘recognition memory’. Nat. Protoc. 1, 1306–1311 (2006).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Research in the laboratory of M.S. is supported by Advanced European Research Council grants (no. 741744), Israel Science Foundation (ISF)-research grant no. 991/16 and ISF-Legacy Heritage Bio-Medical Science Partnership-research grant no. 1354/15. M.S. thanks the Adelis and Thompson Foundations for their generous support of our AD research. I.A. is an incumbent of the Eden and Steven Romick Professorial Chair, supported by Merck KGaA, the Chan Zuckerberg Initiative, the HHMI International Scholar award, the European Research Council Consolidator Grant (ERC-COG, no. 724471- HemTree2.0), an SCA award of the Wolfson Foundation and Family Charitable Trust, the Thompson Family Foundation, an MRA Established Investigator Award (no. 509044), the ISF (no. 703/15), the Ernest and Bonnie Beutler Research Program for Excellence in Genomic Medicine, the Helen and Martin Kimmel award for innovative investigation, the NeuroMac DFG/Transregional Collaborative Research Center Grant, an International Progressive MS Alliance Grant/NMSS (no. PA-1604 08459) and an Adelis Foundation Grant. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. The anti-CCR2 clone (MC21) was created by M. Mack and generously given to M.S.; we thank S. Schwarzbaum for editing the manuscript. Schematic presentations of the experimental design (Figs. 1a,e, 2a, 5a and 6a) were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

R.D.-S., G.C. and M.A. designed, performed and analyzed experiments. Single-cell experiments were designed by R.D.-S., H.K.-S. and A.W. and performed by R.D.-S. and H.K.-S. Data were analyzed by A.W. L.C., T.U., T.C., K.B., C.B. and S.P.C. performed experiments. R.D.-S. and G.C. wrote the manuscript under the supervision of M.S. and I.A., with the help of A.W. and M.A. M.C. generated and provided Trem2−/−5xFAD mice. All experiments were designed, performed and interpreted under the supervision of M.S. and I.A.

Corresponding authors

Correspondence to Assaf Weiner, Ido Amit or Michal Schwartz.

Ethics declarations

Competing interests

M.S. is an inventor of the intellectual property that forms the basis for development of PD-L1 immunotherapy for AD. K.B. is coinventor of the intellectual property that forms the basis for development of PD-L1 immunotherapy for AD. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks Markus Kummer 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.

Extended data

Extended Data Fig. 1 The transcriptomic signature of microglia in Trem2+/+5xFAD mice is not altered upon anti-PD-L1 treatment.

(a) Cumulative bar graphs showing the distribution of homeostatic microglia, stage-I DAM and DAM cells per animal in anti-PD-L1/IgG2b, mouse number, experiment batch and treatment indicated below each bar. (b) Scatter plot comparing DAM cells gene expression from this study and Keren-Shaul et al., 2017 study20, presented as average log2 UMI counts. (c,d) 2D projection plots as in Fig. 1b; highlighting DAM, colored for treatment (anti-PD-L1/IgG2b) (c), or showing the relative expression level of selected marker genes across all microglia (d), color scale indicate the normalized value of log2 fold change. (e) Scatter plots of log2 gene expression (average UMI counts) in homeostatic microglia (i, 1406 cells), stage-I-DAM (ii, 461 cells), and DAM (iii, 1064 cells) derived from the brains of anti-PD-L1 treated and IgG2b-control treated mice. Differential gene expression analysis was performed by Mann-Whitney U test with false-discovery rate (FDR) correction. Red dots represent values that pass the threshold of FDR corrected p < 0.05.

Extended Data Fig. 2 Monocyte-derived macrophages do not contribute to the pool of activated microglia in Trem2+/+5xFAD mice.

(a) FACS gating strategy for isolating all leukocytes (CD45+) and bone marrow (BM)-derived leukocytes (CD45+GFP+) from whole brains of GFP-BM transplanted Trem2+/+5xFAD mice. (b) Cumulative bar graph showing the percentage of each cell population out of all the cells collected using the CD45+ gate or the CD45+GFP+ gate. (c) Cumulative bar graphs of individual animals showing the percentage of each cell population out of all the cells collected using the CD45 + gate or CD45 + GFP + gate in IgG2b or anti-PD-L1 treated animals, corresponding to Fig. 1f,g; also shown are additional 2 WT samples.

Extended Data Fig. 3 Hippocampal levels of Aβ plaques and Triton X-100 Aβ1-42 are not affected by anti-PD-L1 treatment in Trem2-/-5xFAD mice.

(a) Graphic presentation of our RAWM procedure. (b) Graphic presentation of our NOR procedure. (c) RAWM performance of Trem2+/+WT (n=9) and Trem2−/−WT mice (n=12) presented (Y-axis; mean ± SEM) as the number of errors made by the mice (upper panel) and latency to platform (sec, lower panel), in 3-trial bins (x axis) analyzed by two-way ANOVA with repeated measures yielding only main effect of trial bins (upper panel: F(9,171)=45.03, p < 0.0001, 95% CI -1.031-0.8198; lower panel: F(9,171)=66.04, p < 0.0001, 95% CI -6.329-5.767). (d, upper panel) ELISA assessment of Aβ1-40 TBS-soluble hippocampal fractions from anti-PD-L1 (n=9; 7 females (F), 2 males (M)), and IgG2b (n=9; 6 F, 3 M) treated Trem2−/−5xFAD and untreated Trem2−/−WT (n=7; 5 F, 2 M); data derived from 3 cohorts of mice, pooled together after normalization per cohort. (d, lower panel) ELISA assessment of Aβ1-40 Triton X-100-soluble hippocampal fractions from anti-PD-L1 (n=11; 9 F, 2 M) and IgG2b (n=11, 8 F, 3 M) treated Trem2−/−5xFAD and untreated Trem2−/−WT (n=9; 7 F, 2 M); data derived from 4 cohorts of mice, pooled together after normalization per cohort. (e) ELISA assessment of Aβ1-40 TBS-soluble (upper panel) and Triton X-100-soluble (lower panel) hippocampal fractions from anti-PD-L1 (n=7 F) and IgG2b (n=8 F) treated Trem2+/+5xFAD and untreated Trem2+/+WT (n=5 F); data derived from 3 cohorts of mice, pooled together after normalization per cohort. Data were analyzed using one-way ANOVAs followed by a Fisher’s LSD test: (d) TBS-soluble: F(2,22)=25.0, p < 0.0001, R2=0.6944; Triton X-100-soluble: F(2,28)=9.568, p < 0.0001, R2=0.6862; (e) TBS-soluble: F(2,17)=3.479, p=0.0008, R2=0.5653; Triton X-100-soluble: F(2,17)=3.214, p=0.0003, R2=0.623. No significant difference (n.s.) was found following Fisher’s LSD tests. Box plots represent the minimum and maximum values (whiskers), first and third quartile (box boundaries), median (box internal line), and mean (cross); data in all other graphs are shown as mean ± SEM.

Extended Data Fig. 4 No differences in the transcriptomic profile between MDM-1 identified in Trem2−/−5xFAD and Trem2+/+5xFAD and anti-PD-L1 or IgG2b treated mice.

(a) FACS gating strategy for enriching monocyte-derived macrophages (MDM) from the brains of 7-9-month-old Trem2+/+5xFAD and Trem2−/−5xFAD mice, 14 days following injection of anti-PD-L1 or IgG2b. (b) A cumulative bar graphs of individual samples showing the percentage of each cell subpopulation out of all total non-microglial cells collected. (c) A volcano plot showing differentially expressed genes between MDM-1 and DAM, according to MARS-seq data corresponding to Fig. 4. (d) A scatter plot comparing the gene expression profile (log2) of MDM-1 cells collected from the brains of Trem2+/+5xFAD mice (83 cells) and Trem2−/−5xFAD mice (174 cells). (e) A scatter plot comparing the gene expression profile (log2) of MDM-1 cells collected from the brains of mice treated with anti-PD-L1 (83 cells) or IgG2b (102 cells). In figures (c-e) differential gene expression analysis was performed by Mann-Whitney U test with false-discovery rate (FDR) correction.

Extended Data Fig. 5 Anti-PD-L1 antibody target engagement within the peripheral immune system.

(a) Flow cytometry plots demonstrating the gating strategy for T cells (CD3+) from the blood of WT mice. (b) Representative histogram plots of anti-rat IgG2b FITC fluorescent intensity, as a measurement for PD-L1 occupancy, in the saturated tube (‘sat’, orange) and in the tested tube (‘test’, purple) (see ‘Methods’) on CD3+ T cells in blood samples from WT mice, 7 days following treatment with anti-PD-L1 antibody in dose of 0.1, 0.5 or 1.5 mg/mouse, or with 1.5 mg/mouse of IgG2, or untreated (n/group=6; 3 males, 3 females). (c,d) Quantification of the percentage of PD-L1 receptor occupancy (%RO) on T cells (mean ± SEM) from the blood (c) and spleen (d). The %RO is calculated as geometric mean of ‘test’ divided by geometric mean of ‘sat’. (e) Flow cytometry plots demonstrating the gating strategy for PD-1+ effector memory T cells (TEM, CD4+CD44+), measured in the blood of the same mice as detailed above. (f) Percentage of PD-1+ TEM cell out of total TEM (Mean ± SEM). One sample from the group of anti-PD-L1 1.5 mg/mouse was not included for technical reason. (c,d,f) were analyzed using one-way ANOVAs ([c] F(4,25)=7.23, p=0.0005, R2=0.536; [d] F(4,25)=7.16, p=0.0005, R2=0.534; [f] F(4,24)=10.71, p < 0.0001, R2=0.641) followed by Fisher’s LSD tests (n.s. – not significant, ** p < 0.01, *** p < 0.0001). data in all graphs are shown as mean ± SEM.

Extended Data Fig. 6 Blocking CCR2 reduces MDM level in the brain of Trem2-/-5xFAD mice.

(a,b) Locomotor activity measured by total distance that each mouse moved (cm; mean ± SEM) (a), and anxiety measured by time (sec; mean ± SEM) spent in the center of the arena (b), both measured on the habituation phase (day 1) of the NOR task (Trem2−/−WT n=5, Trem2−/−5xFAD/IgG2b n=5, Trem2−/−5xFAD/anti-PD-L1 n=7, Trem2−/−5xFAD/anti-PD-L1 + anti-CCR2 n=8), analyzed using one-way ANOVAs ([a] F(3,21)=0.4152, p=0.744; [b] F(3,21)=0.323, p=0.8). (c) ELISA assessment of hippocampal Aβ1-42 Triton X-100-soluble from anti-PD-L1 (n=7; 5 females, 2 males) and anti-PD-L1+ anti-CCR2 (n=9; 6 females, 3 males) treated Trem2−/−5xFAD mice was analyzed using one-tailed Student’s t test: t(14)=0.126, p=0.45); data derived from 3 cohorts of mice, pooled together after normalization. (d) Flow cytometry plots demonstrating the gating strategy for live single MDM cells in Trem2−/−5xFAD brains. (e) Quantification (presented as mean ± SEM) of MDM (CD45+CD11b+CD44+Ly6G-CD38-) in Trem2−/−5xFAD treated with IgG, anti-PD-L1 or anti-PD-L1 together with anti-CCR2 and untreated Trem2−/−WT; n/group=5. One-way ANOVA yielded a significant main effect (F(3,16)=4.629, p=0.016, R2=0.4647) which was followed by Fisher’s LSD tests. # relates to the comparison between anti-PD-L1 and IgG2b; Φ relates to the comparison with WT; n.s - not significant.,#,Φ indicate p < 0.05.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dvir-Szternfeld, R., Castellani, G., Arad, M. et al. Alzheimer’s disease modification mediated by bone marrow-derived macrophages via a TREM2-independent pathway in mouse model of amyloidosis. Nat Aging 2, 60–73 (2022). https://doi.org/10.1038/s43587-021-00149-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00149-w

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