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:

Sall1 is a transcriptional regulator defining microglia identity and function

An Erratum to this article was published on 19 January 2017

This article has been updated

Abstract

Microglia are the resident macrophages of the central nervous system (CNS). Gene expression profiling has identified Sall1, which encodes a transcriptional regulator, as a microglial signature gene. We found that Sall1 was expressed by microglia but not by other members of the mononuclear phagocyte system or by other CNS-resident cells. Using Sall1 for microglia-specific gene targeting, we found that the cytokine receptor CSF1R was involved in the maintenance of adult microglia and that the receptor for the cytokine TGF-β suppressed activation of microglia. We then used the microglia-specific expression of Sall1 to inducibly inactivate the murine Sall1 locus in vivo, which resulted in the conversion of microglia from resting tissue macrophages into inflammatory phagocytes, leading to altered neurogenesis and disturbed tissue homeostasis. Collectively, our results show that transcriptional regulation by Sall1 maintains microglial identity and physiological properties in the CNS and allows microglia-specific manipulation in vivo.

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

Figure 1: Sall1 expression is restricted mainly to CNS-resident microglia.
Figure 2: CNS-infiltrating myeloid cells and BM-derived microglia and/or macrophages do not express Sall1.
Figure 3: Microglia-specific targeting using Sall1CreER mice.
Figure 4: Gene-expression profile of Sall1-deficient microglia.
Figure 5: Deletion of Sall1 in microglia leads to a reactive phenotype.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

Change history

  • 28 November 2016

    In the HTML version of this article initially published, the scale bar was missing from the inset in the top right image in Figure 2d; the bottom left plot in Figure 2e was incorrectly a duplicate of the adjacent plot at right; and the designations in Figure 4b (Sall1fl and Sall1creER/fl) should have been Sall1fl/fl and Sall1CreER/fl (respectively). Also, the arrows in the designations above and below the plots in Supplementary Figure 3b were rendered as boxes; these should have been as follows: Sall1+/+Cx3cr1CreER-iDTR and Sall1GFP/+Cx3cr1CreER-iDTR. Finally, in Supplementary Figure 4f, the red (Ki67+) cells in the right set of images were not visible. These errors have been corrected for the HTML version of this article.

References

  1. 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 

  2. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hashimoto, D. et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38, 792–804 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518, 547–551 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Hoeffel, G. et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 42, 665–678 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kierdorf, K. et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 16, 273–280 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86–90 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Greter, M. et al. Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia. Immunity 37, 1050–1060 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Wu, Y., Dissing-Olesen, L., MacVicar, B.A. & Stevens, B. Microglia: dynamic mediators of synapse development and plasticity. Trends Immunol. 36, 605–613 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Shemer, A., Erny, D., Jung, S. & Prinz, M. Microglia plasticity during health and disease: an immunological perspective. Trends Immunol. 36, 614–624 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gautier, E.L. et al. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 13, 1118–1128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yamasaki, R. et al. Differential roles of microglia and monocytes in the inflamed central nervous system. J. Exp. Med. 211, 1533–1549 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bennett, M.L. et al. New tools for studying microglia in the mouse and human CNS. Proc. Natl. Acad. Sci. USA 113, E1738–E1746 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Koso, H. et al. Conditional rod photoreceptor ablation reveals Sall1 as a microglial marker and regulator of microglial morphology in the retina. Glia 64, 2005–2024 (2016).

    Article  PubMed  Google Scholar 

  18. Sweetman, D. & Münsterberg, A. The vertebrate spalt genes in development and disease. Dev. Biol. 293, 285–293 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Karantzali, E. et al. Sall1 regulates embryonic stem cell differentiation in association with nanog. J. Biol. Chem. 286, 1037–1045 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Harrison, S.J., Nishinakamura, R., Jones, K.R. & Monaghan, A.P. Sall1 regulates cortical neurogenesis and laminar fate specification in mice: implications for neural abnormalities in Townes-Brocks syndrome. Dis. Model. Mech. 5, 351–365 (2012).

    CAS  PubMed  Google Scholar 

  21. Nishinakamura, R. et al. Murine homolog of SALL1 is essential for ureteric bud invasion in kidney development. Development 128, 3105–3115 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Kohlhase, J. SALL1 mutations in Townes-Brocks syndrome and related disorders. Hum. Mutat. 16, 460–466 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Vodopiutz, J. et al. Homozygous SALL1 mutation causes a novel multiple congenital anomaly-mental retardation syndrome. J. Pediatr. 162, 612–617 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Inoue, S., Inoue, M., Fujimura, S. & Nishinakamura, R. A mouse line expressing Sall1-driven inducible Cre recombinase in the kidney mesenchyme. Genesis 48, 207–212 (2010).

    CAS  PubMed  Google Scholar 

  25. Takasato, M. et al. Identification of kidney mesenchymal genes by a combination of microarray analysis and Sall1-GFP knockin mice. Mech. Dev. 121, 547–557 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Abedin, M.J., Imai, N., Rosenberg, M.E. & Gupta, S. Identification and characterization of Sall1-expressing cells present in the adult mouse kidney. Nephron Exp. Nephrol. 119, e75–e82 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Hirsch, S. et al. A mouse model of Townes-Brocks syndrome expressing a truncated mutant Sall1 protein is protected from acute kidney injury. Am. J. Physiol. Renal Physiol. 309, F852–F863 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Goldmann, T. et al. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat. Immunol. 17, 797–805 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Varvel, N.H. et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc. Natl. Acad. Sci. USA 109, 18150–18155 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bruttger, J. et al. Genetic cell ablation reveals clusters of local self-renewing microglia in the mammalian central nervous system. Immunity 43, 92–106 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Elmore, M.R. 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 

  32. Schreiner, B., Heppner, F.L. & Becher, B. Modeling multiple sclerosis in laboratory animals. Semin. Immunopathol. 31, 479–495 (2009).

    Article  PubMed  Google Scholar 

  33. Croxford, A.L. et al. The Cytokine GM-CSF drives the inflammatory signature of CCR2+ monocytes and licenses autoimmunity. Immunity 43, 502–514 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Waisman, A., Ginhoux, F., Greter, M. & Bruttger, J. Homeostasis of microglia in the adult brain: review of novel microglia depletion systems. Trends Immunol. 36, 625–636 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Remington, L.T., Babcock, A.A., Zehntner, S.P. & Owens, T. Microglial recruitment, activation, and proliferation in response to primary demyelination. Am. J. Pathol. 170, 1713–1724 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Chiu, I.M. et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 4, 385–401 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Goldmann, T. et al. A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat. Neurosci. 16, 1618–1626 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Surace, M.J. & Block, M.L. Targeting microglia-mediated neurotoxicity: the potential of NOX2 inhibitors. Cell. Mol. Life Sci. 69, 2409–2427 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ikushima, H., Negishi, H. & Taniguchi, T. The IRF family transcription factors at the interface of innate and adaptive immune responses. Cold Spring Harb. Symp. Quant. Biol. 78, 105–116 (2013).

    Article  PubMed  Google Scholar 

  41. Ivashkiv, L.B. & Donlin, L.T. Regulation of type I interferon responses. Nat. Rev. Immunol. 14, 36–49 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kimura, A. et al. Aryl hydrocarbon receptor protects against bacterial infection by promoting macrophage survival and reactive oxygen species production. Int. Immunol. 26, 209–220 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Fourgeaud, L. et al. TAM receptors regulate multiple features of microglial physiology. Nature 532, 240–244 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kempermann, G., Jessberger, S., Steiner, B. & Kronenberg, G. Milestones of neuronal development in the adult hippocampus. Trends Neurosci. 27, 447–452 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Ginhoux, F. & Jung, S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14, 392–404 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Gemma, C. & Bachstetter, A.D. The role of microglia in adult hippocampal neurogenesis. Front. Cell. Neurosci. 7, 229 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Ohmori, T., Tanigawa, S., Kaku, Y., Fujimura, S. & Nishinakamura, R. Sall1 in renal stromal progenitors non-cell autonomously restricts the excessive expansion of nephron progenitors. Sci. Rep. 5, 15676 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yamashita, K., Sato, A., Asashima, M., Wang, P.C. & Nishinakamura, R. Mouse homolog of SALL1, a causative gene for Townes-Brocks syndrome, binds to A/T-rich sequences in pericentric heterochromatin via its C-terminal zinc finger domains. Genes Cells 12, 171–182 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Hoeffel, G. et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J. Exp. Med. 209, 1167–1181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Yuri, S. et al. Sall4 is essential for stabilization, but not for pluripotency, of embryonic stem cells by repressing aberrant trophectoderm gene expression. Stem Cells 27, 796–805 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Li, J., Chen, K., Zhu, L. & Pollard, J.W. Conditional deletion of the colony stimulating factor-1 receptor (c-fms proto-oncogene) in mice. Genesis 44, 328–335 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. Levéen, P. et al. TGF-beta type II receptor-deficient thymocytes develop normally but demonstrate increased CD8+ proliferation in vivo. Blood 106, 4234–4240 (2005).

    Article  PubMed  CAS  Google Scholar 

  55. Jung, S. et al. Analysis of fractalkine receptor CX(3)CR1 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 

  56. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat. Methods 2, 419–426 (2005).

    Article  CAS  PubMed  Google Scholar 

  57. Greter, M. et al. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity 36, 1031–1046 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank V. Tosevski and F. Mair for technical help and flow cytometry sorting; J. Jaberg, S. Nemetz and J. Candreia for technical support; A.L. Croxford for critical reading of the manuscript; S. Jessberger, B. Schreiner and C. Raposo for critical discussions; J.K. Georgijevic and W. Qi for performing the next generation sequencing analysis at the Functional Genomics Centre Zurich (FGCZ); Plexxikon (P. Singh and B. West) for PLX5622-containing and control diets; and the Neuroscience Center Zürich, and the Microbiology and Immunology PhD program, University of Zurich and ETH, Zurich, Switzerland. Cx3cr1CreER were kindly provided by S. Jung (Weizmann Institute of Science). Csf1rfl/fl mice were kindly provided by J. Pollard (Albert Einstein College of Medicine). Supported by the Swiss National Science Foundation (BSSGI0_155832, PP00P3_144781, 316030 150768, 310030 146130, and CRSII3 136203 for M.G. and B.B.), the Swiss Multiple Sclerosis Society (M.G. and B.B.), the European Union FP7 project TargetBraIn, NeuroKine, ATECT (B.B.), the National Agency of Research (LIPOCAMD and MACLEAR for E.L.G.) and the Fondation de France (00056835 for E.L.G.).

Author information

Authors and Affiliations

Authors

Contributions

A.B. and M.G. designed the study, performed experiments and wrote the manuscript; R.N. provided the Sall1 strains; M.V., X.Y., N.R.K. and I.L. performed experiments; E.L.G. analyzed microarray data (Immgen); and B.B. designed experiments and co-wrote the manuscript.

Corresponding author

Correspondence to Melanie Greter.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Sall1 expression is restricted to microglia within the hematopoietic system.

(a) Flow cytometry plots show representative pre-gating strategy for CD45+ cells (shown are CNS cells). (b) Flow cytometry analysis of GFP (Sall1) expression in different organs of Sall1GFP/+ and Sall1+/+ (control) mice (pre-gated on CD45+ cells as in a. (c) qPCR analysis of Sall1 mRNA in sorted cell populations derived from WT mice; results were normalized to Pol2 expression. Alveolar MFs: CD45+Siglec-F+CD11c+, Lung CD11b+ DCs: CD45+Siglec-FCD11c+MHCII+CD11b+, Lung CD103+ DCs: CD45+Siglec-FCD11c+MHCII+CD103+, SP MF (spleen macrophages): F4/80hiCD11b+, SP NPs (spleen neutrophils): Ly6G+SSChi, BM Mo (BM monocytes): LinCD11b+Ly6C+CD115+, microglia: CD45loLy6CLy6GCD11b+F4/80+, Per. B cells (peritoneal B cells): B220+, Per. (peritoneal) MFs: CD115+CD11b+F4/80+. (d) qPCR analysis of Sall1 mRNA in total tissue lysates of different organs; results were normalized to Pol2 expression. (e) Representative flow cytometry plots of kidney, liver and heart of Sall1GFP/+ mice (pre-gated on CD45 cells). (f) Quantification of results in e, presented as frequency of GFP+ (Sall1) cells; each symbol represents an individual mouse; small horizontal lines indicate the mean (± s.e.m.). Data are representative of 2-4 mice per genotype, 2 experiments (b); 2 samples per population pooled from 2-3 WT mice, 2 experiments (c; mean ± s.e.m.); 13 (spleen), 12 (brain), 11 (kidney, liver), 9 (spinal cord), 7 (lung, heart), 4 (skin), 3 (lymph node) WT mice, 2-5 experiments, 1 experiment (lymph node) (d; mean ± s.e.m.); 6 (liver), 5 (kidney, heart) Sall1GFP/+ mice, 2 experiments (e,f).

Supplementary Figure 2 Sall1 expression is specific to resident microglia within the adult CNS.

(a) IHC of brain sections of Sall1GFP/+ mice, showing GFP (green), DAPI (blue), and GFAP (radial-glia-like stem cells), DCX (neuroblasts), Calbindin (Purkinje neurons), S100B (astrocytes) or MBP (oligodendrocytes) (red); insets (without DAPI; top right) are enlargements of the outlined areas in the main images. Scale bars, 20 μm (main image) or 5 μm (insets). (b) Gating strategy of non-hematopoietic (CD45) CNS-resident cells and representative flow cytometry plots for their GFP expression in Sall1GFP/+ mice. (c) Quantification of results in b, presented as frequency of GFP+ cells. Each symbol represents an individual mouse; small horizontal lines indicate the mean (± s.e.m.). *P < 0.0001 (one-way ANOVA). (d) Gating strategy of CNS-resident myeloid cells and separately isolated choroid plexus (CP) cells. Representative flow cytometry plots display the percentage of GFP+ cells in Sall1GFP/+ mice. MF: Macrophage. (e) IHC of brain sections of Sall1GFP/+ mice, showing GFP (green), DAPI (blue), Iba-1 or F4/80 (microglia and CNS-MF) (red) and CD31 (endothelial cells) (gray); arrowheads indicate Iba-1 and GFP or F4/80 and GFP double-positive microglia; insets (without DAPI; top right) are enlargements of the outlined areas in the main images. Scale bars, 20 μm (main image) or 5 μm (insets). Data are representative of 2-3 mice per staining, 2 experiments (a); 3 mice, 1 experiment (b,c); 6 mice, 2 experiments (d); 2-3 mice per staining, 2 experiments (e).

Supplementary Figure 3 CNS-infiltrating myeloid cells and BM-derived microglia and/or macrophages do not express Sall1.

(a) Frequency of GFP+ microglia and CD45hi MF of Sall1GFP/+ and Cx3cr1GFP/+ reporter mice and of YFP+ or RFP+ microglia and CD45hi MFs in tamoxifen treated Sall1CreERR26-YFP or Cx3cr1CreERR26-RFP mice. Microglia: CD45loCD11b+F4/80+Ly6CLy6G, CD45hi MFs: CD45hiCD11b+F4/80+Ly6CLy6G. (b) Flow cytometry analysis and quantification of the frequency of GFP+ microglia in tamoxifen- and diphtheria toxin-treated Sall1+/+(CD45.1)→Cx3cr1CreER-iDTR(CD45.2) or Sall1GFP/+(CD45.1)→Cx3cr1CreER-iDTR(CD45.2) BM chimeras on day 0, 11 and 21 after treatment (pre-gated on CD11b+F4/80+CD45.1loLy6CLy6G cells) or in untreated Sall1GFP/+ (control) mice. (c) Representative histograms and quantification of GFP expression in monocyte-derived cells (MCs) (gated on CD45hiCD11b+CD11c+MHCII+Ly6C+), neutrophils (gated on CD45hiCD11b+ Ly6G+), CD4+ T cells (gated on CD45hiCD11b-CD4+) and microglia (gated on CD45loLy6C-Ly6G-CD11b+) in Sall1GFP/+ mice at peak disease of MOG35-55/CFA-induced EAE. Each symbol (a-c) represents an individual mouse; small horizontal lines (a-c) indicate the mean (± s.e.m.). *P < 0.0001 (one-way ANOVA). Data are representative of 15 (Sall1GFP/+), 10 (Sall1CreERR26-YFP, Cx3cr1CreERR26-RFP), 4 (Cx3cr1GFP/+) mice, at least 2 experiments (a); 4 mice (d11, d21), 2 experiments and 1 mouse (d0, untreated Sall1GFP/+) (b); 7 (microglia), 5 (MCs, Neutrophils, CD4+ T cells) mice, 3 experiments (c).

Supplementary Figure 4 Microglia-specific targeting utilizing Sall1CreER mice.

(a) Flow cytometry analysis of Sall1CreERR26-YFP mice and R26-YFP (control) littermates on day 3 after 5 consecutive days of tamoxifen treatment, showing the frequency of YFP+ microglia (pre-gated on CD45loLy6GLy6CCD11b+). (b) IHC of cortical brain sections of Sall1CreERCsf1rfl/fl and Csf1rfl/fl mice at day 0, 7 and 14 after 5 consecutive days of tamoxifen treatment, showing Iba-1 (microglia) (green) and DAPI (blue). Scale bar, 20 μm. Quantification shows microglia counts in different brain areas at day 0 after tamoxifen treatment. (c) qPCR analysis of Tgfbr2 mRNA in microglia sorted from Sall1CreERTgfbr2fl/fl and Tgfbr2fl/fl mice on day 0 after three consecutive days of tamoxifen treatment; results were normalized to Pol2 expression. (d) Histograms display the expression of different surface markers vs. FMO on microglia of Sall1CreERTgfbr2fl/fl and Tgfbr2fl/fl mice as in c on day 6 after tamoxifen treatment. (e) Quantification of microglia numbers in Sall1CreERTgfbr2fl/fl and Tgfbr2fl/fl mice as in c on day 0, 3 and 6 after tamoxifen treatment. Numbers are displayed as ratios to control (Tgfbr2fl/fl) mice. (f) IHC of brain sections from Sall1CreERTgfbr2fl/fl and Tgfbr2fl/fl mice as in c analyzed on day 3 after tamoxifen treatment, showing Iba-1 (green), Ki67 (red) and DAPI (blue). Arrowheads indicate Iba-1 and Ki67 double-positive cells. Scale bar, 50 μm. Each symbol (b,e) represents an individual mouse. ns = not significant, * p < 0.05, ** p < 0.01, *** p < 0.001 (unpaired t-test). Data are representative of 10 mice, 5 experiments (a); 3-5 sections of 2 mice, 2 experiments (b; mean ± s.e.m.); 1 pooled sample of 3-4 mice per genotype (c); 2-5 mice per genotype, at least 2 experiments (d); 4 (d0), 3 (d3), 5-6 (d6) mice, 2 experiments (e; mean ± s.e.m.); 1-2 mice per genotype (f).

Supplementary Figure 5 Gene expression profile of Sall1-deficient microglia.

(a-c) Gene expression analysis of microglia sorted from Sall1CreER/fl and Sall1fl/fl mice on day 1 after 5 times of tamoxifen treatment every second day as described in Figure 4. (a) Venn diagram of differentially expressed genes. (b) Volcano plot showing log2 ratios vs. p values (log10) of all 12,673 detected genes. Genes with highest significance values are annotated. (c) Expression (log2 ratio) of Sall1-regulated genes clustered according to their indicated GO-pathways; IS, Immune system; (bold indicates genes discussed in Results). (d) Multiplex immunoassays show levels (pg/mg) of IL-1, IL-6, TNF-α and IL-10 in serum and whole tissue lysates of spleen, kidney and liver of untreated (control) mice and Sall1CreER/fl and Sall1fl/fl mice at day 6 after start of tamoxifen treatment. (e) Graph displays cell counts of DCX+ neuroblasts in hippocampal brain sections of tamoxifen treated Cx3cr1CreERSall1fl/fl and Cre (control) littermates; each symbol represents an individual mouse; small horizontal lines indicate the mean (± s.e.m.). * p = 0.0006 (unpaired t-test). Data are representative of 3-5 mice pooled per genotype and biological replicate, 3 experiments (a-c); 3 (Sall1CreER/fl, Sall1fl/fl), 2 (untreated) mice, 1 experiment (d; mean ± s.e.m.); 3 (Cx3cr1CreERSall1fl/fl), 4 (control) mice, 2 experiments (e).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 (PDF 4088 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Buttgereit, A., Lelios, I., Yu, X. et al. Sall1 is a transcriptional regulator defining microglia identity and function. Nat Immunol 17, 1397–1406 (2016). https://doi.org/10.1038/ni.3585

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3585

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