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:

Hematopoietic stem cell transplantation chemotherapy causes microglia senescence and peripheral macrophage engraftment in the brain

Abstract

Hematopoietic stem cell transplantation (HSCT) is a therapy used for multiple malignant and nonmalignant diseases, with chemotherapy used for pretransplantation myeloablation. The post-HSCT brain contains peripheral engrafted parenchymal macrophages, despite their absence in the normal brain, with the engraftment mechanism still undefined. Here we show that HSCT chemotherapy broadly disrupts mouse brain regenerative populations, including a permanent loss of adult neurogenesis. Microglial density was halved, causing microglial process expansion, coinciding with indicators of broad senescence. Although microglia expressed cell proliferation markers, they underwent cell cycle arrest in S phase with a majority expressing the senescence and antiapoptotic marker p21. In vivo single-cell tracking of microglia after recovery from chemical depletion showed loss of their regenerative capacity, subsequently replaced with donor macrophages. We propose that HSCT chemotherapy causes microglial senescence with a gradual decrease to a critical microglial density, providing a permissive niche for peripheral macrophage engraftment of the brain.

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: HSCT causes the accumulation of brain parenchymal donor macrophages that become resident and surveillant.
Fig. 2: HSCT busulfan chemotherapy causes loss of host microglia cells and increase in individual microglial process area correlating with donor macrophage density.
Fig. 3: HSCT causes loss of adult neurogenesis and host microglial cell cycle arrest.
Fig. 4: Busulfan chemotherapy causes broad senescence in the brain.
Fig. 5: HSCT host microglial senescence and incapability to recover from PLX3397-induced depletion cause donor macrophage brain parenchymal engraftment.
Fig. 6: Summary of results and busulfan−induced microglial depletion/senescence model of macrophage brain engraftment.

Similar content being viewed by others

Data availability

The data used in the figures for this study are provided in Supplementary information and are covered by the Creative Commons Attribution 4.0 International License (no. CC BY 4.0): https://creativecommons.org/licenses/by/4.0/

References

  1. Kierdorf, K., Katzmarski, N., Haas, C. A. & Prinz, M. Bone marrow cell recruitment to the brain in the absence of irradiation or parabiosis bias. PLoS ONE 8, e58544 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ciurea, S. O. & Andersson, B. S. Busulfan in hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 15, 523–536 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Aubourg, P. et al. Reversal of early neurologic and neuroradiologic manifestations of X-linked adrenoleukodystrophy by bone marrow transplantation. New Engl. J. Med. 322, 1860–1866 (1990).

    Article  CAS  PubMed  Google Scholar 

  4. Cartier, N. et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326, 818–823 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Orchard, P. J. & Tolar, J. Transplant outcomes in leukodystrophies. Semin. Hematol. 47, 70–78 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Shemer, A. et al. Engrafted parenchymal brain macrophages differ from microglia in transcriptome, chromatin landscape and response to challenge. Nat. Commun. 9, 5206 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mildner, A. et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat. Neurosci. 10, 1544–1553 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Peake, K. et al. Bone marrow-derived cell accumulation in the spinal cord is independent of peripheral mobilization in a mouse model of amyotrophic lateral sclerosis. Front. Neurol. 8, 75 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Zhan, L. et al. Proximal recolonization by self-renewing microglia re-establishes microglial homeostasis in the adult mouse brain. PLoS Biol. 17, e3000134 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Böttcher, C., Fernández-Klett, F., Gladow, N., Rolfes, S. & Priller, J. Targeting myeloid cells to the brain using non-myeloablative conditioning. PLoS ONE 8, e80260 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Cronk, J. C. et al. Peripherally derived macrophages can engraft the brain independent of irradiation and maintain an identity distinct from microglia. J. Exp. Med. 215, 1627–1647 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lund, H. et al. Competitive repopulation of an empty microglial niche yields functionally distinct subsets of microglia-like cells. Nat. Commun. 9, 4845 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Peake, K. et al. Busulfan as a myelosuppressive agent for generating stable high-level bone marrow chimerism in mice. J. Vis. Exp. 1, e52553 (2015).

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

    Article  CAS  PubMed  Google Scholar 

  15. Xu, Z. et al. Efficient strategies for microglia replacement in the central nervous system. Cell Rep. 32, 108041 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Meng, A., Wang, Y., Brown, S. A., Zant, G. V. & Zhou, D. Ionizing radiation and busulfan inhibit murine bone marrow cell hematopoietic function via apoptosis-dependent and -independent mechanisms. Exp. Hematol. 31, 1348–1356 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Probin, V., Wang, Y., Bai, A. & Zhou, D. Busulfan selectively induces cellular senescence but not apoptosis in WI38 fibroblasts via a p53-independent but extracellular signal-regulated kinase-p38 mitogen-activated protein kinase-dependent mechanism. J. Pharmacol. Exp. Ther. 319, 551–560 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Iwamoto, T. et al. DNA intrastrand cross-link at the 5’-GA-3’ sequence formed by busulfan and its role in the cytotoxic effect. Cancer Sci. 95, 454–458 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Abdallah, N. M.-B. B., Slomianka, L., Vyssotski, A. L. & Lipp, H.-P. Early age-related changes in adult hippocampal neurogenesis in C57 mice. Neurobiol. Aging 31, 151–161 (2010).

    Article  PubMed  Google Scholar 

  20. Oijen, M. G. C. T., van, Medema, R. H., Slootweg, P. J. & Rijksen, G. Positivity of the proliferation marker Ki-67 in noncycling cells. Am. J. Clin. Pathol. 110, 24–31 (1998).

    Article  PubMed  Google Scholar 

  21. Lombardi, D., Becherucci, F. & Romagnani, P. How much can the tubule regenerate and who does it? An open question. Nephrol. Dial. Transplant. 31, 1243–1250 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Sasaki, K., Matsumura, K., Tsuji, T., Shinozaki, F. & Takahashi, M. Relationship between labeling indices of KI‐67 and brdurd in human malignant tumors. Cancer 62, 989–993 (1988).

    Article  CAS  PubMed  Google Scholar 

  23. Lee, B. Y. et al. Senescence‐associated β‐galactosidase is lysosomal β‐galactosidase. Aging Cell 5, 187–195 (2006).

    Article  CAS  PubMed  Google Scholar 

  24. Freund, A., Laberge, R.-M., Demaria, M. & Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 23, 2066–2075 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Barr, A. R. et al. DNA damage during S-phase mediates the proliferation-quiescence decision in the subsequent G1 via p21 expression. Nat. Commun. 8, 14728 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Huang, Y. et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat. Neurosci. 21, 530–540 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tay, T. L. et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat. Neurosci. 20, 793–803 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Réu, P. et al. The lifespan and turnover of microglia in the human brain. Cell Rep. 20, 779–784 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Askew, K. et al. Coupled proliferation and apoptosis maintain the rapid turnover of microglia in the adult brain. Cell Rep. 18, 391–405 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Campisi, J. & Fagagna, Fd’Addadi Cellular senescence: when bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 8, 729–740 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Oosterhof, N. et al. Colony-stimulating factor 1 receptor (CSF1R) regulates microglia density and distribution, but not microglia differentiation in vivo. Cell Rep. 24, 1203–1217 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Jonas, R. A. et al. The spider effect: morphological and orienting classification of microglia in response to stimuli in vivo. PLoS ONE 7, e30763–12 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lampron, A., Lessard, M. & Rivest, S. Effects of myeloablation, peripheral chimerism, and whole-body irradiation on the entry of bone marrow-derived cells into the brain. Cell Transplant. 21, 1149–1159 (2012).

    Article  PubMed  Google Scholar 

  35. Scholzen, T. & Gerdes, J. The Ki‐67 protein: from the known and the unknown. J. Cell. Physiol. 182, 311–322 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Meng, A., Wang, Y., Zant, G. V. & Zhou, D. Ionizing radiation and busulfan induce premature senescence in murine bone marrow hematopoietic cells. Cancer Res. 63, 5414–5419 (2003).

    CAS  PubMed  Google Scholar 

  37. Jänicke, R. U., Sohn, D., Essmann, F. & Schulze-Osthoff, K. The multiple battles fought by anti-apoptotic p21. Cell Cycle 6, 407–413 (2007).

    Article  PubMed  Google Scholar 

  38. Capotondo, A. et al. Brain conditioning is instrumental for successful microglia reconstitution following hematopoietic stem cell transplantation. Proc. Natl Acad. Sci. USA 109, 15018–15023 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Valley, M. T., Mullen, T. R., Schultz, L. C., Sagdullaev, B. T. & Firestein, S. Ablation of mouse adult neurogenesis alters olfactory bulb structure and olfactory fear conditioning. Front. Neurosci. 3, 51 (2009).

    PubMed  PubMed Central  Google Scholar 

  40. Ghanbari, A. et al. Depletion of neural stem cells from the subventricular zone of adult mouse brain using cytosine b‐arabinofuranoside. Brain Behav. 5, e00404 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Egeland, M. et al. Depletion of adult neurogenesis using the chemotherapy drug temozolomide in mice induces behavioural and biological changes relevant to depression. Transl. Psychiatry 7, e1101 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Buchbinder, D. et al. Neurocognitive dysfunction in hematopoietic cell transplant recipients: expert review from the late effects and Quality of Life Working Committee of the CIBMTR and complications and Quality of Life Working Party of the EBMT. Bone Marrow Transplant. 53, 535–555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wolf, N. I. et al. Metachromatic leukodystrophy and transplantation: remyelination, no cross‐correction. Ann. Clin. Transl. Neurol. 7, 169–180 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double‐fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Jung, S. et al. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell Biol. 20, 4106–4114 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Holtmaat, A. et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nat. Protoc. 4, 1128–1144 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sailor, K. A. et al. Persistent structural plasticity optimizes sensory information processing in the olfactory bulb. Neuron 91, 384–396 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sun, G. J. et al. Seamless reconstruction of intact adult-born neurons by serial end-block imaging reveals complex axonal guidance and development in the adult hippocampus. J. Neurosci. 33, 11400–11411 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank P. Aubourg for his inspiration to initiate this study; J. Strauch, I. Bernstein and G. Dufayet-Chaffaud for experimental support; E. Gomez Perdiguero, G. Lepousez, I. Gabanyi, S. Voytek, F. Koukouli, H. Song, G. Eberl, L. Peduto, F. Jagot-Brunner, G. Parsons and A. Giniatullina for advice; and T. Sailor for two-photon stage fabrication. This work was partly funded by bluebird bio, Inc. as part of a collaboration (K.A.S., P.-M.L. and N.C.), Agence Nationale de la Recherche (no. ANR-15-CE37-0004-01, to K.A.S. and P.-M.L.), Agence Nationale de la Recherche (no. ANR-15-NEUC-0004, Circuit-OPL, to K.A.S. and P.-M.L.), the life insurance company ’AG2R-La-Mondiale’ (to K.A.S. and P.-M.L.), the Agence Nationale de la Recherche ‘Investissements d’avenir’ program (no. ANR-19P3IA-0001, PRAIRIE 31A Institute, to C.G., J.-B.M. and C.L.V.) and a pre- and postdoctoral fellowship from the Laboratory for Excellence (LabEx) ‘Revive’ (no. ANR-10-LABX-73, to C.G. and K.A.S.). S.L.-M, S.T., B.G.-L. and N.C. funding from the program “Investissements d’avenir” ANR-10-IAIHU-06 and ANR-11-INBS-0011–NeurATRIS: Translational Research Infrastructure for Biotherapies in Neurosciences. Part of this work was carried out in the PHENOPARC core facility, CELIS core facility and ICAN core facility of ICM.

Author information

Authors and Affiliations

Authors

Contributions

K.A.S. undertook conceptualization, investigation, methodology and writing of the original draft. G.A. carried out conceptualization, investigation, methodology, writing review and editing. S.L.-M., B.G.-L. and S.T. were responsible for conceptualization and investigation. C.G. and C.L.V. carried out methodology. J.-B.M. performed methodology and supervision. M.B. and K.G. were responsible for conceptualization, writing of the review and editing. G.V. undertook conceptualization. P.-M.L. carried out supervision, conceptualization, writing of the review and editing. N.C. carried out supervision, conceptualization, writing of the review and editing.

Corresponding authors

Correspondence to Kurt A. Sailor, Pierre-Marie Lledo or Nathalie Cartier.

Ethics declarations

Competing interests

This project was partially funded by bluebird bio, Inc. for reagents and supplies, with partial salaries provided to G.A., B.G.-L., S.L.-M. and S.T.; M.B. and K.G. are current employees, and G.V. a former employee, of bluebird bio, Inc.; K.A.S., C.G., J.-B.M., C.L.V., P.-M.L. and N.C. declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Joanne Kurtzberg and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Jerome Staal was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team

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 HSCT donor cell macrophage characteristics within brain.

(a) Plot of donor cells that are negative for macrophage or microglia markers (tdTom+/Iba-1-/TMEM119; mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; ***P < 0.001, **P < 0.01, *P < 0.05; P = 0.0025, 0.0019, 0.0006 and 0.0128 for 2, 4, 6 12 and 24 weeks, respectively, vs. 24 weeks; n = 4 mice for 2 and 4 weeks, n = 6 mice for 6, 12 and 24 weeks). (b) Plot of density of donor cells (tdTom+) in brain regions of olfactory bulb (OB), hippocampus, dorsal cerebral cortex (DorCtx), lateral cerebral cortex (LatCtx) and striatum at 6, 12 and 24 weeks post-HSCT. The OB has a significantly higher density of cells compared to all other brain regions at 12 and 24 weeks. (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; P < 0.0001 for OB 12 and 24 weeks vs. 6 weeks, ****P < 0.0001 for non-OB regions vs. OB at respective week with **P = 0.0095 24 weeks VentCtx vs. OB.; n = 3, 4 and 3 mice for 6, 12 and 24 weeks post-HSCT, respectively). (c) Surface renderings of three example donor cells at 17, 19 and 23 weeks post-HSCT. Scale bar = 15 µm. (d) Cell surface area plot of the same donor cells in vivo imaged from 12 to 25 weeks post-HSCT. (Light red plot of individual cells, black plot mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; n = 5, 10, 11, 9, 11, 11, 9, 7 cells for 12, 15.3, 16.3, 17.4, 18.9, 19.1, 23.1 and 24.4 weeks post-HSCT, respectively).

Extended Data Fig. 2 Host microglia density decline with HSCT, donor and host macrophage properties.

(a) The same registered brain region 2-photon in vivo imaged. Scale bar = 100 µm. (b) Microglia density comparing between in vivo 2-photon imaged CX3CR1-GFP mice and histology (tdTom/Iba-1+/TMEM119+) analysis. (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; n = 4 animals all groups). (c) Immunohistochemistry at 24 weeks showing donor macrophages (tdTom+/Iba-1+/TMEM119, white closed arrowhead), host microglia (tdTom/Iba-1+/TMEM119+, white open arrowhead) and host macrophages (tdTom/Iba-1+/TMEM119, blue open arrowhead). Scale bar = 20 μm. (d) Plot of host macrophage density at control (Ctrl), 1 day (1d), 1, 2, 4, 6, 12 and 24 weeks post-HSCT (mean ± SEM; two-sided Kruskal-Wallis with Dunn’s multiple comparisons test; **P < 0.01; P = 0.0063 12 weeks vs. Ctrl; n = 7, 7, 4, 4, 4, 6, 6 and 6 mice for Ctrl, 1d, 1, 2, 4, 6, 12 and 24 weeks, respectively). (e) Plot of donor macrophage density vs. host microglia density at 12 and 24 weeks post-HSCT. (Mean of individual brain sections; simple linear regression [black and orange dashed lines] significance from slope = 0, P = 0.5382 F = 0.3930 R2 = 0.02026 and P = 0.0007 F = 13.86 R2 = 0.2958 for 12 and 24 weeks, respectively; n = 24 and 35 brain sections for 12 and 24 weeks, respectively). (f) Macrophage convex hull plot at 12 and 24 weeks post-HSCT (mean ± SEM; two-sided unpaired t-test; n = 5 and 6 mice for 12 and 24 weeks post-HSCT, respectively). (g) Convex hull plot of donor and host parenchymal macrophages (mean ± SEM; two-sided unpaired t-test; n = 1, 5 and 6 mice for 6, 12 and 24 week donor cells post-HSCT, respectively [binned], and n = 4 and 6 mice for 12 and 24 week host cells post-HSCT, respectively [binned]).

Extended Data Fig. 3 Effect of HSCT on SVZ proliferation, adult neurogenesis neural progenitors and derived immature neurons.

(a) lateral ventricle (LV) sub-ventricular zone with immunohistochemistry for Ki67+ cells (open arrowhead). Scale bar = 100 μm. (b) Plot of Ki67+ density in subventricular zone of the lateral ventricles (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; ****P < 0.0001 Ctrl vs. all; n = 4, 3, 4, 4, 4, 6, 6 and 3 mice for Ctrl, 1d, 1, 2, 4, 6, 12 and 24 weeks, respectively). (c) Adult hippocampus dentate gyrus region showing doublecortin (DCX) positive adult neurogenesis neural progenitor cells/immature neurons in the subgranule zone (open arrowhead). Scale bar = 100 µm. (d) Adult olfactory bulb in control (Ctrl) doublecortin labeling (open arrowhead). Scale bar = 200 µm. (e) Plot of doublecortin adult neurogenesis neural progenitor cell density in dentate gyrus subgranule zone of hippocampus with orange circles representing modeled normal age-related decline of adult neurogenesis DCX. (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; **** p < 0.0001 Ctrl vs. 6, 12 and 24 weeks; n = 4, 3, 4, 3 mice for Ctrl, 6, 12 and 24 weeks, respectively). (f) Plot of doublecortin relative fluorescent intensity (% max control) in sub-ventricular zone of lateral ventricles (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; **** p < 0.0001 Ctrl vs. 6, 12 and 24 weeks; n = 4, 3, 4, 3 mice for Ctrl, 6, 12 and 24 weeks, respectively). (g) Plot of doublecortin relative fluorescent intensity (%max control) in olfactory bulb (mean ± SEM; two-sided unpaired t-test; ***P = 0.0001 Ctrl vs. 24 weeks; n = 4, 3 mice for Ctrl and 24 weeks, respectively).

Extended Data Fig. 4 Ventral to dorsal brain pattern of NG2 depletion and partial recovery with HSCT.

(a) Confocal images of NG2 immunohistochemistry of dorsal cortex in control (Ctrl), 4 and 24 weeks post-HSCT with NG2+ cells indicated (open arrowhead). Scale bar = 50 µm. (b) Plot of NG2+ cell density at control (Ctrl), 1 day, 1, 2, 4, 6, 12 and 24 weeks post-HSCT (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; ****P < 0.0001 for 1 to 12 weeks vs. Ctrl and **P = 0.0021 for 24 weeks vs. Ctrl; n = 7, 3, 4, 4, 4, 6, 6, and 3 mice for Ctrl, 1d, 1, 2, 4, 6, 12, 24 weeks post-HSCT, respectively). (c) Tiled brain hemispheres at Control, 2, 4, 6, 12 and 24 weeks post-HSCT of NG2 cells. Control brain section has dense labeling of NG2 cells throughout cortex. Two weeks post-HSCT most NG2 cells are lost in cortex but small population of dense labeled NG2 cells found in thalamic regions (open arrowhead). By four weeks the section is devoid of NG2 cells. For 6, 12 and 24 weeks the NG2 cells returned but with a ventral reconstitution and a clear demarcation (dashed black line) from dorsal cortical regions. This pattern was observed in most mice, with a few individual outliers having dorsal near-complete recovery (Extended Data Fig. 4b at 6-12 weeks). Scale bar = 1 mm.

Extended Data Fig. 5 Effect of HSCT on donor cell proliferation and host microglia Ki67 modeling.

(a) Plot of donor macrophage proliferation at 2, 4, 6, 12 and 24 weeks post-HSCT (mean ± SEM; one-way two-sided ANOVA with Tukey multiple comparisons test; n = 4, 4, 6, 6, and 3 for 2, 4, 6, 12, and 24 weeks post-HSCT, respectively). (b) Plot of microglia density (green dashed line and spots; from Fig. 2a) with density model (orange line) at a 5%/day loss rate from Ctrl to 2 weeks post-HSCT and 10%/day from 2-4 weeks post-HSCT as an adjustment for inferred Ki67 increased proliferation. Using this 10%/day loss rate, there would be a complete depletion of host microglia by 12 weeks (orange dotted-line) due to loss of assumed Ki67 driven cell proliferation.

Extended Data Fig. 6 Single cell tracking of in vivo 2-photon image microglia.

Scatter plot above of total number of microglia in a FOV for each mouse, with color matched to tracks below, during PLX recovery period. Bottom plot of individual cell tracks (horizontal colored lines) for individual mice from HSCT group for 9 days of PLX3397 recovery. Each line represents one tracked cell and its survival duration.

Extended Data Fig. 7 Donor cell re-population of brain with effective loss of host microglia, proliferation of donor cells and migration front.

(a) In vivo 2-photon projected image of mouse 1 with successful donor cell engraftment after PLX-recovery and mouse 2, unsuccessful donor cell engraftment after PLX-recovery showing higher density of host microglia. Scale bar = 50 µm. (b) In vivo detected mitotic spheres of proliferating cells segregated into days after PLX3397 recovery from Fig. 4f (mean ± SEM; n = 5 mice). (c) Migration front at 0 min (red dashed line) and 120 min (green dashed line) at 6d post-PLX3397 showing migration direction (white arrows). Scale bar = 50 µm. (d) Same FOV (green dashed line) as in c, at 6, 7 and 8 days post-PLX. Scale bar = 100 µm.

Extended Data Fig. 8 PLX/busulfan efficient replacement of brain microglia population with donor macrophages and effect on donor and host motility and cortical neuron density.

(a) Tiled images of histological sections at 21 weeks post-HSCT of mice from PLX recovery experiment (Fig. 4) showing pattern of tdTom (red) and CX3CR1-GFP (green) cells having an average of 91.27 ± 3.67 % replacement of brain area with donor macrophages. Mouse 5 FOVs highlight typical morphology of host microglia (FOV 1) with large cell area and complex, fine process pattern compared to host peripheral macrophages (FOV 2) having a smaller cell area with less complex process pattern. FOV 3 demonstrates efficient white matter engraftment of donor macrophages. Scale bar for whole-brain images = 500 µm and for FOV images scale bar = 20 µm. (b) Plot of process area change (%/10 min) between control, no HSCT microglia (Ctrl microglia), sparse post-HSCT host microglia (sparse HSCT microglia), post-HSCT/post-PLX macrophages (dense donor macrophages) and post-HSCT macrophages (sparse donor macrophages). (mean ± SD; one-way two-sided ANOVA with Tukey multiple comparisons test; *P = 0.0408 Ctrl microglia vs. sparse HSCT microglia, ***P = 0.0002 sparse HSCT microglia vs. dense donor macrophages, and ****P < 0.0001 for dense donor macrophages and control microglia vs. sparse donor macrophages; n = 40, 28, 40 and 20 cells for Ctrl microglia, sparse HSCT microglia, dense donor macrophages and sparse donor macrophages, respectively). (c) Plot of NeuN+ cell density of layer 2/3 in dorsal cortex between control (Ctrl) and 21 weeks post-HSCT, PLX withdrawal donor macrophage enriched (HSCT) brains. (mean ± SEM; two-sided unpaired t-test; p = 0.9780).

Supplementary information

Reporting Summary

Supplementary Video 1

Donor cell tdTom+ peripherally derived meningeal phagocytes and intravascular cells. Two-photon in vivo imaged time-lapse video at 1 week post HSCT showing initially detected tdTom+ cells within the meningeal space as phagocytes, imaged at 10-s intervals (arrowhead in first half of video). Blood vessels show a continuous stream of circulating tdTom cells, as well as cells adhering to the lumen and migrating (arrowhead in second half of video).

Supplementary Video 2

Donor cell tdTom+ macrophages in brain parenchyma. Two-photon in vivo imaged z-stack movie (superficial to deep) at 2 months post HSCT showing initially detected tdTom+ macrophages in brain parenchyma (arrowhead), with depth from brain surface indicated. Also note high density of tdTom+ blood cells within the vasculature.

Supplementary Video 3

Sparse donor cell-derived tdTom+ macrophage process dynamics. Two-photon in vivo imaged time-lapse video of tdTom+ macrophage process dynamics imaged at 10-min intervals for 50 min. Left panel shows raw images, and right highlights new (green) and lost (red) processes.

Supplementary Video 4

Donor macrophage cell division within brain parenchyma and cell detection within vasculature. Two-photon in vivo imaged time-lapse video imaged at 10-min intervals for 2 h of tdTom+ macrophages (purple heatmap) undergoing massive cell division and migration at 6 days recovery from PLX3397 treatment, with blood vessels labeled in red. The first half of the video highlights mitotic spheres (arrowhead) and division events while the second half shows an example of a donor blood cell within a blood vessel for attempted tracking of transmigration events.

Supplementary Video 5

Donor macrophage migration front. Two-photon in vivo imaged time-lapse video of tdTom+ macrophage (purple heatmap) migration front at edge of brain region largely devoid of microglia/macrophages. The first half of the video highlights examples of mitotic spheres (arrowhead) behind the migration front, and the second half is at more accelerated speed with a line highlighting the edge of the migration front.

Supplementary Data 1

Data used in study figures.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sailor, K.A., Agoranos, G., López-Manzaneda, S. et al. Hematopoietic stem cell transplantation chemotherapy causes microglia senescence and peripheral macrophage engraftment in the brain. Nat Med 28, 517–527 (2022). https://doi.org/10.1038/s41591-022-01691-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-01691-9

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