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.

  • Resource
  • Published:

Mutations in the human CSF1R gene impact microglia’s maintenance of brain white matter integrity

Abstract

Microglia, the brain’s resident macrophages, depend on interleukin-34 and colony-stimulating factor 1 (CSF1) for their development and maintenance, engaging the CSF1 receptor (CSF1R). Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), a neurodegenerative disorder affecting the brain’s white matter, is caused by heterozygous pathogenic mutations in the CSF1R gene. This study investigated molecular mechanisms underlying ALSP using single-nucleus RNA sequencing on postmortem brain specimens. Results showed a significant reduction in microglia in ALSP brains, with remaining cells exhibiting a unique activation signature. This reduction correlated with decreased myelinating oligodendrocytes (OLs) and increased neuropilin-2+ OLs with a stress-response and anti-apoptotic signature, driven by STAT3 and fibroblast growth factor receptor pathways. Additionally, astrocytes displayed maladaptive activation and stress responses. These findings underscore microglia’s crucial role in supporting OL myelination and limiting astrocyte repair responses, suggesting therapeutic strategies balancing CSF1R, fibroblast growth factor receptor and STAT3 pathways for ALSP and other genetically caused microgliopathies.

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: Remaining microglia in individuals with ALSP exhibit signatures of activation and lipid metabolism.
Fig. 2: ALSP OLs display a unique signature.
Fig. 3: ALSP and Csf1rΔFIRE/ΔFIRE OLs are transcriptionally similar.
Fig. 4: ALSP astrocytes exhibit a stress-response signature.
Fig. 5: Spatial transcriptomic analysis corroborates the molecular signatures enriched in ALSP white matter.

Similar content being viewed by others

Data availability

All requests for human snRNA-seq data are promptly reviewed by the University Hospital Leipzig, Children’s Hospital of Philadelphia and Brain Research Institute at Niigata University to verify if the request is subject to any intellectual property or confidentiality obligations. Human-related data not included in the paper were generated as part of clinical trials and may be subject to patient confidentiality. Any data and materials that can be shared will be released via a material transfer agreement. The snRNA-seq data generated in this study are available in the Gene Expression Omnibus under accession number GSE295854. The spatial transcriptomic data generated in this study are uploaded to Zenodo and available at: https://doi.org/10.5281/zenodo.15186190. The following datasets were previously published and reanalyzed in this study: snRNA-seq of occipital cortex from control individuals (under accession number GSE190013), snRNA-seq from three control white matter (under accession number GSE180759), and scRNA-seq from Csf1rΔFIRE/ΔFIRE mice (under accession number GSE267545).

Code availability

The code used to analyze scRNA-seq are available upon request.

References

  1. Borst, K., Dumas, A. A. & Prinz, M. Microglia: immune and non-immune functions. Immunity 54, 2194–2208 (2021).

    CAS  PubMed  Google Scholar 

  2. Paolicelli, R. C. et al. Microglia states and nomenclature: a field at its crossroads. Neuron 110, 3458–3483 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Paolicelli, R. C. et al. Synaptic pruning by microglia is necessary for normal brain development. Science 333, 1456–1458 (2011).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Tremblay, M.-È., Lowery, R. L. & Majewska, A. K. Microglial interactions with synapses are modulated by visual experience. PLoS Biol. 8, e1000527 (2010).

    PubMed  PubMed Central  Google Scholar 

  6. Sierra, A. et al. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell 7, 483–495 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Badimon, A. et al. Negative feedback control of neuronal activity by microglia. Nature 586, 417–423 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Hoffmann, A. & Miron, V. E. CNS macrophage contributions to myelin health. Immunol. Rev. 327, 53–70 (2024).

    CAS  PubMed  Google Scholar 

  9. Kent, S. A. & Miron, V. E. Microglia regulation of central nervous system myelin health and regeneration. Nat. Rev. Immunol. 24, 49–63 (2024).

    CAS  PubMed  Google Scholar 

  10. Irfan, M., Evonuk, K. S. & DeSilva, T. M. Microglia phagocytose oligodendrocyte progenitor cells and synapses during early postnatal development: implications for white versus gray matter maturation. FEBS J. 289, 2110–2127 (2022).

    CAS  PubMed  Google Scholar 

  11. Nemes-Baran, A. D., White, D. R. & DeSilva, T. M. Fractalkine-dependent microglial pruning of viable oligodendrocyte progenitor cells regulates myelination. Cell Rep. 32, 108047 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Wlodarczyk, A. et al. A novel microglial subset plays a key role in myelinogenesis in developing brain. EMBO J. 36, 3292–3308 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Hagemeyer, N. et al. Microglia contribute to normal myelinogenesis and to oligodendrocyte progenitor maintenance during adulthood. Acta Neuropathol. 134, 441–458 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Papapetropoulos, S. et al. Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia: review of clinical manifestations as foundations for therapeutic development. Front. Neurol. 12, 788168 (2021).

    PubMed  Google Scholar 

  15. Konno, T. et al. Clinical and genetic characterization of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia associated with CSF1R mutation. Eur. J. Neurol. 24, 37–45 (2017).

    CAS  PubMed  Google Scholar 

  16. Papapetropoulos, S. et al. Clinical presentation and diagnosis of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia: a literature analysis of case studies. Front. Neurol. 15, 1320663 (2024).

    PubMed  PubMed Central  Google Scholar 

  17. Muthusamy, K. et al. Adult-onset leukodystrophies: a practical guide, recent treatment updates, and future directions. Front. Neurol. 14, 1219324 (2023).

    PubMed  PubMed Central  Google Scholar 

  18. Rademakers, R. et al. Mutations in the colony stimulating factor 1 receptor (CSF1R) gene cause hereditary diffuse leukoencephalopathy with spheroids. Nat. Genet. 44, 200–205 (2011).

    PubMed  PubMed Central  Google Scholar 

  19. Konno, T., Kasanuki, K., Ikeuchi, T., Dickson, D. W. & Wszolek, Z. K. CSF1R-related leukoencephalopathy: a major player in primary microgliopathies. Neurology 91, 1092–1104 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Dulski, J., Sundal, C. & Wszolek, Z. K. CSF1R-related disorder. in GeneReviews (eds. M. P. Adam et al.) (University of Washington, 1993).

  21. Dulski, J., Muthusamy, K., Lund, T. C. & Wszolek, Z. K. CSF1R-related disorder: state of the art, challenges, and proposition of a new terminology. Parkinsonism Relat. Disord. 121, 105894 (2024).

    CAS  PubMed  Google Scholar 

  22. Hume, D. A. et al. Phenotypic impacts of CSF1R deficiencies in humans and model organisms. J. Leukoc. Biol. 107, 205–219 (2020).

    CAS  PubMed  Google Scholar 

  23. Stanley, E. R. & Chitu, V. CSF-1 receptor signaling in myeloid cells. Cold Spring Harb. Perspect. Biol. 6, a021857–a021857 (2014).

    PubMed  PubMed Central  Google Scholar 

  24. Stanley, E. R., Biundo, F., Gökhan, Ş. & Chitu, V. Differential regulation of microglial states by colony stimulating factors. Front. Cell Neurosci. 17, 1275935 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang, Y. et al. IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia. Nat. Immunol. 13, 753–760 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Pixley, F. J. & Stanley, E. R. CSF-1 regulation of the wandering macrophage: complexity in action. Trends Cell Biol. 14, 628–638 (2004).

    CAS  PubMed  Google Scholar 

  28. Chitu, V., Gökhan, Ş. & Stanley, E. R. Modeling CSF-1 receptor deficiency diseases - how close are we? FEBS J. 289, 5049–5073 (2022).

    CAS  PubMed  Google Scholar 

  29. Wu, J. et al. The phenotypic and genotypic spectrum of CSF1R-related disorder in China. Mov. Disord. 39, 798–813 (2024).

    CAS  PubMed  Google Scholar 

  30. NCBI MedGEn. Brain abnormalities, neurodegeneration, and dysosteosclerosis (concept ID: C5193117); https://www.ncbi.nlm.nih.gov/medgen/C5193117/

  31. Guo, L. & Ikegawa, S. From HDLS to BANDDOS: fast-expanding phenotypic spectrum of disorders caused by mutations in CSF1R. J. Hum. Genet. 66, 1139–1144 (2021).

    CAS  PubMed  Google Scholar 

  32. Tada, M. et al. Characteristic microglial features in patients with hereditary diffuse leukoencephalopathy with spheroids. Ann. Neurol. 80, 554–565 (2016).

    CAS  PubMed  Google Scholar 

  33. 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).

    CAS  PubMed  Google Scholar 

  34. Konno, T. et al. Haploinsufficiency of CSF-1R and clinicopathologic characterization in patients with HDLS. Neurology 82, 139–148 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Berdowski, W. M. et al. Dominant-acting CSF1R variants cause microglial depletion and altered astrocytic phenotype in zebrafish and adult-onset leukodystrophy. Acta Neuropathol. 144, 211–239 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Siletti, K. et al. Transcriptomic diversity of cell types across the adult human brain. Science 382, eadd7046 (2023).

    CAS  PubMed  Google Scholar 

  38. Dikmen, H. O. et al. GM-CSF induces noninflammatory proliferation of microglia and disturbs electrical neuronal network rhythms in situ. J. Neuroinflammation 17, 235 (2020).

    PubMed  PubMed Central  Google Scholar 

  39. Yamasaki, R. et al. Restoration of microglial function by granulocyte-colony stimulating factor in ALS model mice. J. Neuroimmunol. 229, 51–62 (2010).

    CAS  PubMed  Google Scholar 

  40. Basso, L. et al. Granulocyte-colony-stimulating factor (G-CSF) signaling in spinal microglia drives visceral sensitization following colitis. Proc. Natl Acad. Sci. USA 114, 11235–11240 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, Y. et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat. Med. 26, 131–142 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mathys, H. et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570, 332–337 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Absinta, M. et al. A lymphocyte–microglia–astrocyte axis in chronic active multiple sclerosis. Nature 597, 709–714 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Masuda, T. et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 566, 388–392 (2019).

    CAS  PubMed  Google Scholar 

  45. Marschallinger, J. et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat. Neurosci. 23, 194–208 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Haney, M. S. et al. APOE4/4 is linked to damaging lipid droplets in Alzheimer’s disease microglia. Nature 628, 154–161 (2024).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. De Schepper, S. et al. Perivascular cells induce microglial phagocytic states and synaptic engulfment via SPP1 in mouse models of Alzheimer’s disease. Nat. Neurosci. 26, 406–415 (2023).

    PubMed  PubMed Central  Google Scholar 

  48. Lei, S., Hu, M. & Wei, Z. Single-cell sequencing reveals an important role of SPP1 and microglial activation in age-related macular degeneration. Front. Cell Neurosci. 17, 1322451 (2023).

    CAS  PubMed  Google Scholar 

  49. Pandey, S. et al. Disease-associated oligodendrocyte responses across neurodegenerative diseases. Cell Rep. 40, 111189 (2022).

    CAS  PubMed  Google Scholar 

  50. Sadick, J. S. et al. Astrocytes and oligodendrocytes undergo subtype-specific transcriptional changes in Alzheimer’s disease. Neuron 110, 1788–1805 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Xiang, X., Zhang, X. & Huang, Q. -L. Plexin A3 is involved in semaphorin 3F-mediated oligodendrocyte precursor cell migration. Neurosci. Lett. 530, 127–132 (2012).

    CAS  PubMed  Google Scholar 

  52. Piaton, G. et al. Class 3 semaphorins influence oligodendrocyte precursor recruitment and remyelination in adult central nervous system. Brain 134, 1156–1167 (2011).

    PubMed  Google Scholar 

  53. Williams, A. et al. Semaphorin 3A and 3F: key players in myelin repair in multiple sclerosis? Brain 130, 2554–2565 (2007).

    PubMed  Google Scholar 

  54. Kaminski, W. E., Wenzel, J. J., Piehler, A., Langmann, T. & Schmitz, G. ABCA6, a novel a subclass ABC transporter. Biochem. Biophys. Res. Commun. 285, 1295–1301 (2001).

    CAS  PubMed  Google Scholar 

  55. Soane, L., Rus, H., Niculescu, F. & Shin, M. L. Inhibition of oligodendrocyte apoptosis by sublytic C5b-9 is associated with enhanced synthesis of bcl-2 and mediated by inhibition of caspase-3 activation. J. Immunol. 163, 6132–6138 (1999).

    CAS  PubMed  Google Scholar 

  56. Kögel, D., Linder, B., Brunschweiger, A., Chines, S. & Behl, C. At the crossroads of apoptosis and autophagy: multiple roles of the co-chaperone BAG3 in stress and therapy resistance of cancer. Cells 9, 574 (2020).

    PubMed  PubMed Central  Google Scholar 

  57. McCluggage, F. & Fox, A. H. Paraspeckle nuclear condensates: global sensors of cell stress? Bioessays 43, e2000245 (2021).

    PubMed  Google Scholar 

  58. Dzwonek, J. & Wilczynski, G. M. CD44: molecular interactions, signaling and functions in the nervous system. Front. Cell Neurosci. 9, 175 (2015).

    PubMed  PubMed Central  Google Scholar 

  59. Tuohy, T. M. F. et al. CD44 overexpression by oligodendrocytes: a novel mouse model of inflammation-independent demyelination and dysmyelination. Glia 47, 335–345 (2004).

    PubMed  Google Scholar 

  60. Steelman, A. J. et al. Activation of oligodendroglial Stat3 is required for efficient remyelination. Neurobiol. Dis. 91, 336–346 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).

    CAS  PubMed  Google Scholar 

  63. Tian, Y. et al. CXCL12 induces migration of oligodendrocyte precursor cells through the CXCR4‑activated MEK/ERK and PI3K/AKT pathways. Mol. Med. Rep. 18, 4374–4380 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Kang, M. & Yao, Y. Laminin regulates oligodendrocyte development and myelination. Glia 70, 414–429 (2022).

    CAS  PubMed  Google Scholar 

  65. Mierzwa, A. J., Zhou, Y. -X., Hibbits, N., Vana, A. C. & Armstrong, R. C. FGF2 and FGFR1 signaling regulate functional recovery following cuprizone demyelination. Neurosci. Lett. 548, 280–285 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Rajendran, R., Giraldo-Velásquez, M., Stadelmann, C. & Berghoff, M. Oligodendroglial fibroblast growth factor receptor 1 gene targeting protects mice from experimental autoimmune encephalomyelitis through ERK/AKT phosphorylation. Brain Pathol. 28, 212–224 (2018).

    CAS  PubMed  Google Scholar 

  67. Rajendran, R. et al. Oligodendrocyte-specific deletion of FGFR1 reduces cerebellar inflammation and neurodegeneration in MOG35–55-induced EAE. Int. J. Mol. Sci. 22, 9495 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Kamali, S. et al. Oligodendrocyte-specific deletion of FGFR2 ameliorates MOG35–55-induced EAE through ERK and Akt signalling. Brain Pathol. 31, 297–311 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Bansal, R. & Pfeiffer, S. E. FGF-2 converts mature oligodendrocytes to a novel phenotype. J. Neurosci. Res. 50, 215–228 (1997).

    CAS  PubMed  Google Scholar 

  70. Thümmler, K. et al. Polarizing receptor activation dissociates fibroblast growth factor 2 mediated inhibition of myelination from its neuroprotective potential. Acta Neuropathol. Commun. 7, 212 (2019).

    PubMed  PubMed Central  Google Scholar 

  71. Jin, S., Plikus, M. V. & Nie, Q. CellChat for systematic analysis of cell–cell communication from single-cell transcriptomics. Nat. Protoc. 20, 180–219 (2025).

    CAS  PubMed  Google Scholar 

  72. Chadarevian, J. P. et al. Therapeutic potential of human microglia transplantation in a chimeric model of CSF1R-related leukoencephalopathy. Neuron https://doi.org/10.1016/j.neuron.2024.05.023 (2024).

  73. Munro, D. A. D. et al. Microglia protect against age-associated brain pathologies. Neuron https://doi.org/10.1016/j.neuron.2024.05.018 (2024).

  74. McNamara, N. B. et al. Microglia regulate central nervous system myelin growth and integrity. Nature https://doi.org/10.1038/s41586-022-05534-y (2022).

  75. González-Velasco, Ó., Sanchez-Luis, E., Rosa, E. D. L., Sánchez-Santos, J. M. & Rivas, J. D. L. A single-cell clusters similarity measure for different batches, datasets, and samples. Preprint at bioRxiv https://doi.org/10.1101/2022.03.14.483731 (2022).

  76. Rothstein, J. D. et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron 16, 675–686 (1996).

    CAS  PubMed  Google Scholar 

  77. Yi, J. -H. & Hazell, A. S. Excitotoxic mechanisms and the role of astrocytic glutamate transporters in traumatic brain injury. Neurochem. Int. 48, 394–403 (2006).

    CAS  PubMed  Google Scholar 

  78. Zukerberg, L. R. et al. Loss of cables, a cyclin-dependent kinase regulatory protein, is associated with the development of endometrial hyperplasia and endometrial cancer. Cancer Res. 64, 202–208 (2004).

    CAS  PubMed  Google Scholar 

  79. Ohta, Y., Hartwig, J. H. & Stossel, T. P. FilGAP, a Rho- and ROCK-regulated GAP for Rac binds filamin A to control actin remodelling. Nat. Cell Biol. 8, 803–814 (2006).

    CAS  PubMed  Google Scholar 

  80. Rothhammer, V. et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 22, 586–597 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Hou, J. et al. Transcriptomic atlas and interaction networks of brain cells in mouse CNS demyelination and remyelination. Cell Rep. 42, 112293 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Jacobo-Albavera, L., Domínguez-Pérez, M., Medina-Leyte, D. J., González-Garrido, A. & Villarreal-Molina, T. The role of the ATP-binding cassette A1 (ABCA1) in human disease. Int. J. Mol. Sci. 22, 1593 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Karasinska, J. M. et al. Specific loss of brain ABCA1 increases brain cholesterol uptake and influences neuronal structure and function. J. Neurosci. 29, 3579–3589 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Wheeler, M. A. et al. Environmental control of astrocyte pathogenic activities in CNS inflammation. Cell 176, 581–596 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Clark, I. C. et al. Identification of astrocyte regulators by nucleic acid cytometry. Nature 614, 326–333 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Liu, C. -C. et al. Cell-autonomous effects of APOE4 in restricting microglial response in brain homeostasis and Alzheimer’s disease. Nat. Immunol. 24, 1854–1866 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Fields, J. & Ghorpade, A. C/EBPβ regulates multiple IL-1β-induced human astrocyte inflammatory genes. J. Neuroinflammation 9, 177 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Shaftel, S. S., Griffin, W. S. T. & O’Banion, M. K. The role of interleukin-1 in neuroinflammation and Alzheimer disease: an evolving perspective. J. Neuroinflammation 5, 7 (2008).

    PubMed  PubMed Central  Google Scholar 

  89. Ali, Z. S., Van Der Voorn, J. P. & Powers, J. M. A comparative morphologic analysis of adult onset leukodystrophy with neuroaxonal spheroids and pigmented glia-a role for oxidative damage. J. Neuropathol. Exp. Neurol. 66, 660–672 (2007).

    PubMed  Google Scholar 

  90. Alturkustani, M., Keith, J., Hazrati, L. -N., Rademakers, R. & Ang, L. -C. Pathologic staging of white matter lesions in adult-onset leukoencephalopathy/leukodystrophy with axonal spheroids. J. Neuropathol. Exp. Neurol. 74, 233–240 (2015).

    CAS  PubMed  Google Scholar 

  91. Chen, Y. et al. The functions and roles of sestrins in regulating human diseases. Cell. Mol. Biol. Lett. 27, 2 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Pereira, B. I. et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat. Immunol. 21, 684–694 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Phipson, B. et al. Propeller: testing for differences in cell type proportions in single cell data. Bioinformatics 38, 4720–4726 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Teng, X. -Y. et al. OPALIN is an LGI1 receptor promoting oligodendrocyte differentiation. Proc. Natl Acad. Sci. USA 121, e2403652121 (2024).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Kenigsbuch, M. et al. A shared disease-associated oligodendrocyte signature among multiple CNS pathologies. Nat. Neurosci. 25, 876–886 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Pan, J. et al. Deciphering glial contributions to CSF1R-related disorder via single-nuclear transcriptomic profiling: a case study. Acta Neuropathol. Commun. 12, 139 (2024).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Safaiyan, S. et al. Age-related myelin degradation burdens the clearance function of microglia during aging. Nat. Neurosci. 19, 995–998 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Cantuti-Castelvetri, L. et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science 359, 684–688 (2018).

    CAS  PubMed  Google Scholar 

  99. Pasquini, L. A. et al. Galectin-3 drives oligodendrocyte differentiation to control myelin integrity and function. Cell Death Differ. 18, 1746–1756 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Thomas, L. & Pasquini, L. A. Galectin-3-mediated glial crosstalk drives oligodendrocyte differentiation and (re)myelination. Front. Cell Neurosci. 12, 297 (2018).

    PubMed  PubMed Central  Google Scholar 

  101. de Jong, C. G. H. M., Gabius, H. -J. & Baron, W. The emerging role of galectins in (re)myelination and its potential for developing new approaches to treat multiple sclerosis. Cell. Mol. Life Sci. 77, 1289–1317 (2020).

    PubMed  Google Scholar 

  102. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Zhou, Y. et al. Human early-onset dementia caused by DAP12 deficiency reveals a unique signature of dysregulated microglia. Nat. Immunol. 24, 545–557 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Paloneva, J., Autti, T., Hakola, P. & Haltia, M. J. Polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy. in GeneReviews (eds. M. P. Adam et al.) (University of Washington, 1993).

  105. Chitu, V. et al. Phenotypic characterization of a Csf1r haploinsufficient mouse model of adult-onset leukodystrophy with axonal spheroids and pigmented glia (ALSP). Neurobiol. Dis. 74, 219–228 (2015).

    CAS  PubMed  Google Scholar 

  106. Chitu, V. et al. Microglial homeostasis requires balanced CSF-1/CSF-2 receptor signaling. Cell Rep. 30, 3004–3019 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Stables, J. et al. A kinase-dead Csf1r mutation associated with adult-onset leukoencephalopathy has a dominant inhibitory impact on CSF1R signalling. Development 149, dev200237 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Dulski, J. et al. Hematopoietic stem cell transplantation in CSF1R-related leukoencephalopathy: retrospective study on predictors of outcomes. Pharmaceutics 14, 2778 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Colonna, M. The biology of TREM receptors. Nat. Rev. Immunol. 23, 580–594 (2023).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank C. Mirescu (Vigil), S. Brioschi (Washington University in St. Louis) and J. P. Chadarevian (University of California, Irvine) for critical reading and helpful suggestions; C. Chen (Washington University in St. Louis) for providing Oil Red O reagent; A. Watson and the Anatomic and Molecular Pathology Core Labs (Washington University in St. Louis) for helping with histology. We also thank J. Hou (Washington University in St. Louis), J. Han (Washington University in St. Louis) and Z. Miao (University of Pennsylvania) for insightful discussions on transcriptomic analysis. We appreciate the support from L. Spears and M. Castillo (10x Genomics), as well as J. Ponce and the GTAC core (Washington University in St. Louis), for their assistance with spatial profiling. This work was supported by National Institutes of Health grants RF1 AG051485, R21 AG059176 and P01 AG078106; the Cure Alzheimer’s Fund; and Fred and Ginger Haberle Charitable Fund at East Texas Communities Foundation to M. Colonna, and Grant SPP2395 from the German Research Foundation to C.G.B. The Global Leukodystrophy Initiative Clinical Trials Network Pilot Project Career Development Award to J.L.O.-M. M.T. was supported by the Promotion of Science (JSPS, 22H02995). A.K. is supported by the Strategic Research Program for Brain Science from AMED (JP23wm0425019) and the Collaborative Research Project of the Brain Research Institute, Niigata University, Japan.

Author information

Authors and Affiliations

Authors

Contributions

S.D., Y.Z. and M. Colonna designed the study and interpreted the results. S.D. and Y.Z. prepared single-nuclei samples and performed computational analyses. D.L., M. Cella, H.H., J.W., J.L., Z.C. and J.K. assisted with the analyses. C.G.B., J.L.O.-M., M.T. and A.K. provided human postmortem brain samples and clinical advice. S.D., Y.Z., J.K. and M. Colonna wrote the manuscript with feedback from the authors.

Corresponding author

Correspondence to Marco Colonna.

Ethics declarations

Competing interests

M. Colonna is a member of the scientific advisory board of Vigil, NGMBio, Cartesian, Cell Signaling Technology and Halyard; receives research support from Vigil and Ono Pharmaceutical; and has patents pending on TREM2 and LILRB4. J.L.O.-M. is a site principal investigator for Vigil Neuroscience clinical trials. J.K. is cofounder of Rho Bio. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks David Hume, Clare Pridans and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: S. Houston, in collaboration with the Nature Immunology 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 Quality controls and cluster characterization of brain cells in control and ALSP samples.

a. Bar graphs of median of number of genes and median of number of UMIs of each sample sequenced in each batch. b. UMAP plot of control and ALSP samples before batch correction. c-e. UMAP plots of control and ALSP samples after batch correction by Harmony. UMAP plots are split by different samples, genders and regions respectively. f. Heatmap showing the expression of marker genes for each cell type. g. Nuclei frequencies for all cell types in each sample. For the brain regions, frontal cortex (FC), occipital cortex (OC), subcortical regions (SC), parietal and temporal lobes, basal ganglia (BG), and white matter (WM) were assessed. h. UMAP plots depicting the expression of feature genes in different cell types. i. Box plots showing average individual expression levels for CSF1R, CSF2RA, CSF3R in the myeloid cell cluster. P values were calculated by two-sided Wilcoxon rank-sum test (n = 13 controls; n = 6 ALSP). j. Scatter plot showing linear regression of average expression levels between CSF1R and CSF2RA, or CSF3R. Gray shading represents the 95% confidence interval, and Pearson’s correlation coefficient and P value are shown.

Extended Data Fig. 2 Cluster characterization of the myeloid cell subsets in control and ALSP samples.

a. UMAP plots of expression of feature genes in different myeloid clusters (re-cluster from the Micro cluster in Fig. 1a). b. Heatmap showing the average gene expression of the top upregulated microglia genes in ALSP versus healthy control samples. c. Box plots showing average individual expression levels for selected DEGs in the myeloid cell cluster. P values were calculated by two-sided Wilcoxon rank-sum test (n = 13 controls; n = 6 ALSP). d. UMAP plot of gene module score of genes related to ferroptosis pathway (Wikipathway: WP4313) (left panel); Violin plot of FTL expression in myeloid cells between the conditions (right panel). P values were determined by two-sided Wilcoxon rank-sum test. e. UMAP plots of control and ALSP brain myeloid cells grouped by different brain regions. For the brain regions, frontal cortex (FC), occipital cortex (OC), subcortical regions (SC), parietal and temporal lobes, basal ganglia (BG), and white matter (WM) were analyzed. f-h. Violin plots displaying the gene set scores of DEGs derived from two AD datasets (f), two MS datasets (g), and LDAM dataset (h) in control and ALSP myeloid cells. The gene set scores were calculated by UCell. P values were determined by two-sided Wilcoxon rank-sum test. Box center lines, bounds of boxes and whiskers indicate median, first and third quartiles and minima and maxima within a 1.5× IQR, respectively. i. Representative images of Oil Red O, CD163 and Hematoxylin staining in the white matter of control and ALSP brain samples. j-k. Scatter plot showing linear regression of average expression levels between CSF1R and selected genes, revealing they are positively (j) or negatively (k) correlated with CSF1R. Gray shading represents the 95% confidence interval; Pearson’s correlation coefficient and P value are shown.

Extended Data Fig. 3 Integration of white matter data with a public dataset, and cluster characterization of myeloid cell subsets in control and ALSP samples.

a. Representative IHC images of CD163 staining in control and ALSP brain samples (left), with corresponding quantification (right). Each data point represents the value from a single image. For each individual, 5 images were taken and analyzed. Statistical significance was assessed using a non-parametric t-test. Data are shown as box-and-whisker plots (min to max) with all data points displayed. Images from individual donors (n = 3 controls; n = 2 ALSP) are denoted by distinct shapes (circle, triangle, or rectangle). Scale bar = 100 µm. b. Representative IHC images of LGALS3 staining in control and ALSP brain samples (left), with corresponding quantification (right). Each data point represents the value from a single image. For each individual, 5 images were taken and analyzed. Statistical significance was assessed using a non-parametric t-test. Data are shown as box-and-whisker plots (min to max) with all data points displayed. Images (n = 15 controls; n = 10 ALSP) from individual donors (n = 3 controls; n = 2 ALSP) are denoted by distinct shapes (circle, triangle, or rectangle). Scale bar = 100 µm. c. UMAP plot of nuclei from the white matter (WM) snRNA data from Absinta et al., and the current study before integration. d. UMAP plot of nuclei isolated from the white matter brains of controls and individuals with ALSP after integration; n = 39,624 total nuclei; n = 5 controls and n = 4 ALSP patients. Ex_neuron, excitatory neurons; In_neuron, inhibitory neurons; Oligo, oligodendrocytes; OPC, oligodendrocyte precursor cells; Astro, astrocytes; Micro, microglia; Endo, endothelial cells; Stroma, stromal cells; IMM, immune cells. e. UMAP plots of nuclei isolated from the white matter brains of controls and individuals with ALSP after integration, grouped by individuals. In this analysis, nuclei from the current study (Control-1 and Control-4) were renamed as Control-1 and Control-2, respectively; nuclei from three control white matter samples from Absinta et al. (CTRL1, CTRL2, CTRL3) were renamed as Control-3, Control-4, and Control-5. f. UMAP plots of control and ALSP white matter brain macrophages. g. Nuclei frequency of each myeloid cell subsets across all samples. h. Venn diagrams of overlapping genes commonly upregulated in ALSP microglia from the white matter and ALSP microglia from all regions.

Extended Data Fig. 4 Cluster characterization of OLs in control and ALSP samples.

a. Dot plot showing expression of marker genes for each OL subset. b. Heatmap showing the gene set scores of the OL subsets from Pandey et al. and Sadick et al., on the OL subset identified in the current study. c. Nuclei frequency of each OL subset across all samples. d. Venn diagrams of overlapping genes commonly upregulated in ALSP OLs and signature genes of Oligo4 subset. e. Heatmap showing the average gene expression of the top upregulated genes in ALSP versus control OLs. f. Violin plots displaying the gene set scores of ALSP, AD and MS OLs, and MAPK signaling pathway in control and ALSP OLs. The gene set scores were calculated by UCell. P values were determined by two-sided Wilcoxon rank-sum test. Box center lines, bounds of boxes and whiskers indicate median, first and third quartiles and minima and maxima within a 1.5× IQR, respectively. g. Representative images of IHC staining of MBP (i), NF200 (ii), TPPP (iii), GFAP (iv) in ALSP brain sections; the grey matter (GM) and white matter (WM) are indicated. Scale bar = 500 µm except for iii where is 50 µm. Arrowheads highlight OLs with strong expression of TPPP/p25 in the subcortex (left) and weak to no expression in the progressing lesion (middle to right) on the same tissue section. h. Dot plot showing expression of marker genes for each subset of OL-lineage cells. i. IPA analysis of OLs identifies FGF2 as a significant upstream regulator activated in ALSP OLs. P-value was determined using Fisher’s exact test. j. Representative immunofluorescent images of GFAP and FGF2 staining in control and ALSP brain samples (left), with corresponding quantification of FGF2⁺ nuclei per image (right). Statistical significance was assessed using a non-parametric t-test. Each data point represents the value from a single image. Statistical significance was assessed using a non-parametric t-test. Data are shown as box-and-whisker plots (min to max) with all data points displayed. Images (n = 15 controls; n = 10 ALSP) from individual donors (n = 3 controls; n = 2 ALSP) are denoted by distinct shapes (circle, triangle, diamond, or rectangle). Scale bar = 100 µm.

Extended Data Fig. 5 Cluster characterization of white matter OLs in control and ALSP samples.

a. UMAP plot of white matter OLs (re-cluster from the Oligo cluster in Extended Data Fig. 3d) from the integrated dataset. b. Donut plots showing nuclei distribution of each OL subset in control or ALSP samples. c. Nuclei frequency of each OL subsets across all samples. d. Venn diagrams of overlapping genes commonly upregulated in ALSP OLs from the white matter and ALSP OLs from all regions. e. Pan-cell type relative information flow showing signaling pathways identified in ALSP (in red) and control (blue) white matter cells from the CellChat analysis. f. Chord diagrams plotting FGF signaling strength with the senders and receivers with their predicted ligands and receptors from the CellChat analysis. The lines represent changes in ligand-receptor interaction strength. g. Circle plot showing directed interaction of selected signaling pathways between cell types from the CellChat analysis.

Extended Data Fig. 6 Cluster characterization of astrocytes in control and ALSP samples.

a. Representative IHC images showing GFAP staining in the frontal cortex of control and ALSP brain samples (left), with corresponding quantification (right). Statistical significance was determined using a non-parametric t-test. Data are presented as box-and-whisker plots with all data points; n = 8 control, n = 3 ALSP. Scale bar = 20 µm. b. Dot plot showing expression of marker genes for each astrocyte subset. c. Nuclei frequencies of each astrocyte subset across all samples. d. Heatmap showing the average gene expression of the top upregulated genes in ALSP versus control astrocytes. e. Molecular Function (MF) pathways enriched in ALSP OLs. f. Box plots showing average individual expression levels for selected DEGs in astrocytes. P values were calculated by two-sided Wilcoxon rank-sum test. g. UMAP plots of expression of IL1B across all cell types. h. Representative IHC images showing ASC staining in early-stage (left) and advanced-stage (right) ALSP white matter lesions. Increased ASC signal is observed in advanced lesions, indicating enhanced inflammasome activation. Scale bar = 50 µm.

Extended Data Fig. 7 Cluster characterization of white matter astrocytes in control and ALSP samples.

a. UMAP plot of the integrated dataset of white matter cells from ALSP and control brains. b. UMAP plot of astrocytes (re-cluster from the Astro cluster in Extended Data Fig. 3d) from the integrated dataset. c. Nuclei frequencies of each astrocyte subset across all samples. d. Dot plot showing expression of marker genes for each astrocyte subset. e. Volcano plot depicting DEGs in ALSP versus control astrocytes in the white matter. f. GO-MF enriched in astrocytes from ALSP individuals. g. GO-MF enriched in astrocytes from controls in comparison to ALSP.

Extended Data Fig. 8 Cluster characterization of endothelial, stromal, and immune cells in control and ALSP samples.

a. Volcano plot depicting DEGs in ALSP versus control capillary blood endothelial cells (cBEC). b. Molecular function (MF) enriched in cBEC from ALSP (upper panel) or control (lower panel) individuals. c. IPA pathways predicted to be activated (upper panel) or inhibited (lower panel) in ALSP cBEC. d. UMAP plot of stromal cells re-cluster from the Stroma and Peri_Fibro clusters. e. Dot plot showing expression of marker genes for each stromal subset. f. Volcano plot depicting DEGs in ALSP versus control pericytes. g. Molecular Function (MF) enriched in perivascular fibroblasts from ALSP compared to controls. h. IPA of pathways predicted to be activated in ALSP pericytes. i. UMAP plots of immune cells re-cluster from the IMM clusters. j. Dot plot showing expression of marker genes for each immune cell subset. k. UMAP plots of immune cells from different individuals. l. Violin plots displaying the expression of selected genes in control and ALSP immune cells.

Extended Data Fig. 9 Xenium in situ spatial characterization in control and ALSP white matter.

a. Representative hematoxylin-stained images and immunofluorescent images of control (left) and ALSP (right) white matter samples used for spatial transcriptomic profiling. The cell boundary staining, aSMA/Vimentin and DAPI staining were performed in the Xenium multimodal Cell Segmentation Staining workflow. Scale bar = 1000 μm. b. Heatmap showing the expression of marker genes for each cell type. c. Dot plot showing expression of marker genes for each OL subset. d. Spatial maps of control (left) and ALSP white matter (right) samples showing MOG and ST18 expression. Scale bar = 1000 μm. e. Spatial maps of control (left) and ALSP white matter (right) samples showing BAG3, SERPINA3, IFITM3 expression. Scale bar = 1000 μm. f. Spatial maps of control (left) and ALSP (right) white matter samples displaying module scores for the glial cell differentiation gene ontogeny pathway. Differences in spatial distribution suggest altered glial maturation in ALSP. Scale bar = 1000 μm. g. Spatial maps of control (left) and ALSP (right) white matter samples showing module scores for the regulation of response to stress gene ontogeny pathway. Increased stress-related transcriptional signatures are evident in ALSP white matter. Scale bar = 1000 μm.

Supplementary information

Supplementary Information

Supplementary Table 1 Participant information and CSF1R mutations carried by individuals assessed in the study. Supplementary mutation information and references.

Reporting Summary

Supplementary Table 2

DEGs of ALSP versus control in each cell type.

Supplementary Table 3

Raw data used for IHC and immunofluorescence quantification.

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

Du, S., Zhou, Y., Li, D. et al. Mutations in the human CSF1R gene impact microglia’s maintenance of brain white matter integrity. Nat Immunol 26, 1198–1211 (2025). https://doi.org/10.1038/s41590-025-02195-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-025-02195-7

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