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:

Diet-dependent regulation of TGFβ impairs reparative innate immune responses after demyelination

An Author Correction to this article was published on 23 February 2023

This article has been updated

Abstract

Proregenerative responses are required for the restoration of nervous-system functionality in demyelinating diseases such as multiple sclerosis (MS). Yet, the limiting factors responsible for poor CNS repair are only partially understood. Here, we test the impact of a Western diet (WD) on phagocyte function in a mouse model of demyelinating injury that requires microglial innate immune function for a regenerative response to occur. We find that WD feeding triggers an ageing-related, dysfunctional metabolic response that is associated with impaired myelin-debris clearance in microglia, thereby impairing lesion recovery after demyelination. Mechanistically, we detect enhanced transforming growth factor beta (TGFβ) signalling, which suppresses the activation of the liver X receptor (LXR)-regulated genes involved in cholesterol efflux, thereby inhibiting phagocytic clearance of myelin and cholesterol. Blocking TGFβ or promoting triggering receptor expressed on myeloid cells 2 (TREM2) activity restores microglia responsiveness and myelin-debris clearance after demyelinating injury. Thus, we have identified a druggable microglial immune checkpoint mechanism regulating the microglial response to injury that promotes remyelination.

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: WD impairs lesion recovery after demyelinating injury.
Fig. 2: Correcting metabolic alterations induced by WD improves lesion recovery after demyelination.
Fig. 3: Demyelinated lesions of WD-fed mice accumulate myelin- and crystal-loaded phagocytes.
Fig. 4: The LXR pathway is insufficiently activated in WD-fed mice.
Fig. 5: Impaired microglia activation correlates with increased TGFβ signalling in the brain after WD feeding.
Fig. 6: TGFβ blocks cholesterol efflux gene induction after myelin uptake.
Fig. 7: Blocking TGFβ signalling in microglia of WD-fed mice promotes recovery from demyelinating injury.
Fig. 8: Treatment with TREM2-activating monoclonal antibodies 4D9 improves lipid clearance in phagocytes after demyelinating injury.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request. Source data for the lipidomics analyses and uncropped images of western blots are provided. Source data are provided with this paper.

Code availability

The script used for lesion volume analysis can be found at: https://github.com/lenkavaculciakova/lesion_volume

Change history

References

  1. Hotamisligil, G. S. Inflammation, metaflammation and immunometabolic disorders. Nature 542, 177–185 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Hansson, G. K. & Hermansson, A. The immune system in atherosclerosis. Nat. Immunol. 12, 204–212 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Chawla, A., Nguyen, K. D. & Goh, Y. P. Macrophage-mediated inflammation in metabolic disease. Nat. Rev. Immunol. 11, 738–749 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lloyd, A. F. & Miron, V. E. The pro-remyelination properties of microglia in the central nervous system. Nat. Rev. Neurol. https://doi.org/10.1038/s41582-019-0184-2 (2019).

  5. Franklin, R. J. M. & Ffrench-Constant, C. Regenerating CNS myelin—from mechanisms to experimental medicines. Nat. Rev. Neurosci. 18, 753–769 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Plemel, J. R., Liu, W. Q. & Yong, V. W. Remyelination therapies: a new direction and challenge in multiple sclerosis. Nat. Rev. Drug Discov. 16, 617–634 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Lampron, A. et al. Inefficient clearance of myelin debris by microglia impairs remyelinating processes. J. Exp. Med. 212, 481–495 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kotter, M. R., Li, W. W., Zhao, C. & Franklin, R. J. Myelin impairs CNS remyelination by inhibiting oligodendrocyte precursor cell differentiation. J. Neurosci. 26, 328–332 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Prinz, M., Jung, S. & Priller, J. Microglia biology: one century of evolving concepts. Cell 179, 292–311 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Stadelmann, C., Timmler, S., Barrantes-Freer, A. & Simons, M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol. Rev. 99, 1381–1431 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Miron, V. E. et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat. Neurosci. 16, 1211–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dombrowski, Y. et al. Regulatory T cells promote myelin regeneration in the central nervous system. Nat. Neurosci. 20, 674–680 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yeung, M. S. Y. et al. Dynamics of oligodendrocyte generation in multiple sclerosis. Nature 566, 538–542 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jakel, S. et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature https://doi.org/10.1038/s41586-019-0903-2 (2019).

  15. Kuhlmann, T. et al. Differentiation block of oligodendroglial progenitor cells as a cause for remyelination failure in chronic multiple sclerosis. Brain 131, 1749–1758 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Patrikios, P. et al. Remyelination is extensive in a subset of multiple sclerosis patients. Brain 129, 3165–3172 (2006).

    Article  PubMed  Google Scholar 

  17. Chang, A., Tourtellotte, W. W., Rudick, R. & Trapp, B. D. Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N. Engl. J. Med. 346, 165–173 (2002).

    Article  PubMed  Google Scholar 

  18. Reich, D. S., Lucchinetti, C. F. & Calabresi, P. A. Multiple sclerosis. N. Engl. J. Med. 378, 169–180 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Baecher-Allan, C., Kaskow, B. J. & Weiner, H. L. Multiple sclerosis: mechanisms and immunotherapy. Neuron 97, 742–768 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Neumann, B. et al. Metformin restores CNS remyelination capacity by rejuvenating aged stem cells. Cell Stem Cell 25, 473–485 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cantuti-Castelvetri, L. et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science https://doi.org/10.1126/science.aan4183 (2018).

  22. Ruckh, J. M. et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 10, 96–103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shen, S. et al. Age-dependent epigenetic control of differentiation inhibitors is critical for remyelination efficiency. Nat. Neurosci. 11, 1024–1034 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sim, F. J., Zhao, C., Penderis, J. & Franklin, R. J. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J. Neurosci. 22, 2451–2459 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gianfrancesco, M. A. & Barcellos, L. F. Obesity and multiple sclerosis susceptibility: a review. J. Neurol. Neuromedicine 1, 1–5 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Munger, K. L., Chitnis, T. & Ascherio, A. Body size and risk of MS in two cohorts of US women. Neurology 73, 1543–1550 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Dalby, M. J., Ross, A. W., Walker, A. W. & Morgan, P. J. Dietary uncoupling of gut microbiota and energy harvesting from obesity and glucose tolerance in mice. Cell Rep. 21, 1521–1533 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jeffery, N. D. & Blakemore, W. F. Remyelination of mouse spinal cord axons demyelinated by local injection of lysolecithin. J. Neurocytol. 24, 775–781 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Plemel, J. R. et al. Microglia response following acute demyelination is heterogeneous and limits infiltrating macrophage dispersion. Sci. Adv. 6, eaay6324 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lloyd, A. F. et al. Central nervous system regeneration is driven by microglia necroptosis and repopulation. Nat. Neurosci. 22, 1046–1052 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, Y. et al. Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–432 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Evans, R. M. & Mangelsdorf, D. J. Nuclear receptors, RXR, and the Big Bang. Cell 157, 255–266 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Huang, J. K. et al. Retinoid X receptor gamma signaling accelerates CNS remyelination. Nat. Neurosci. 14, 45–53 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Cunha, M. I. et al. Pro-inflammatory activation following demyelination is required for myelin clearance and oligodendrogenesis. J. Exp. Med 217, e20191390 (2020).

  35. Bogie, J. F. J. et al. Stearoyl-CoA desaturase-1 impairs the reparative properties of macrophages and microglia in the brain. J. Exp. Med. 217, e20191660 (2020).

  36. Fielding, C. J. & Fielding, P. E. Molecular physiology of reverse cholesterol transport. J. Lipid Res. 36, 211–228 (1995).

    Article  CAS  PubMed  Google Scholar 

  37. Bogie, J. F. et al. Myelin-derived lipids modulate macrophage activity by liver X receptor activation. PLoS ONE 7, e44998 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Spann, N. J. et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell 151, 138–152 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Schroepfer, G. J. Jr. Oxysterols: modulators of cholesterol metabolism and other processes. Physiol. Rev. 80, 361–554 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Yan, J. et al. Obesity- and aging-induced excess of central transforming growth factor-beta potentiates diabetic development via an RNA stress response. Nat. Med. 20, 1001–1008 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Samad, F., Yamamoto, K., Pandey, M. & Loskutoff, D. J. Elevated expression of transforming growth factor-beta in adipose tissue from obese mice. Mol Med 3, 37–48 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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 

  43. Wrana, J. L., Attisano, L., Wieser, R., Ventura, F. & Massague, J. Mechanism of activation of the TGF-β receptor. Nature 370, 341–347 (1994).

    Article  CAS  PubMed  Google Scholar 

  44. Zoller, T. et al. Silencing of TGFβ signalling in microglia results in impaired homeostasis. Nat. Commun. 9, 4011 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Seystahl, K., Wick, W. & Weller, M. Therapeutic options in recurrent glioblastoma–an update. Crit. Rev. Oncol. Hematol. 99, 389–408 (2016).

    Article  PubMed  Google Scholar 

  46. Song, W. M. & Colonna, M. The identity and function of microglia in neurodegeneration. Nat. Immunol. 19, 1048–1058 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Cignarella, F. et al. TREM2 activation on microglia promotes myelin debris clearance and remyelination in a model of multiple sclerosis. Acta Neuropathol. https://doi.org/10.1007/s00401-020-02193-z (2020).

  48. Deczkowska, A., Amit, I. & Schwartz, M. Microglial immune checkpoint mechanisms. Nat. Neurosci. 21, 779–786 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Jaitin, D. A. et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell 178, 686–698 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Nugent, A. A. et al. TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron https://doi.org/10.1016/j.neuron.2019.12.007 (2019).

  51. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).

    Article  CAS  PubMed  Google Scholar 

  52. Kleinberger, G. et al. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci. Transl. Med. 6, 243ra286 (2014).

    Article  Google Scholar 

  53. Wunderlich, P. et al. Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and gamma-secretase-dependent intramembranous cleavage. J. Biol. Chem. 288, 33027–33036 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Schlepckow, K. et al. Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region. EMBO Mol. Med. 12, e11227 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kalin, S. et al. Hypothalamic innate immune reaction in obesity. Nat. Rev. Endocrinol. 11, 339–351 (2015).

    Article  PubMed  Google Scholar 

  56. Madore, C., Yin, Z., Leibowitz, J. & Butovsky, O. Microglia, lifestyle stress, and neurodegeneration. Immunity https://doi.org/10.1016/j.immuni.2019.12.003 (2020).

  57. Guillemot-Legris, O. & Muccioli, G. G. Obesity-induced neuroinflammation: beyond the hypothalamus. Trends Neurosci. 40, 237–253 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Prinz, M. & Priller, J. Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat. Rev. Neurosci. 15, 300–312 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Deczkowska, A. et al. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell 173, 1073–1081 (2018).

    Article  CAS  PubMed  Google Scholar 

  60. Krasemann, S. et al. The TREM2–APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566–581 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Grajchen, E., Hendriks, J. J. A. & Bogie, J. F. J. The physiology of foamy phagocytes in multiple sclerosis. Acta Neuropathol. Commun. 6, 124 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Endo, F. et al. Astrocyte-derived TGF-β1 accelerates disease progression in ALS mice by interfering with the neuroprotective functions of microglia and T cells. Cell Rep. 11, 592–604 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. Baror, R. et al. Transforming growth factor-beta renders ageing microglia inhibitory to oligodendrocyte generation by CNS progenitors. Glia 67, 1374–1384 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Xu, J. et al. TGF-β in mice ameliorates experimental autoimmune encephalomyelitis in regulating NK cell activity. Cell Transplant. 28, 1155–1160 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hamaguchi, M. et al. Circulating transforming growth factor-β1 facilitates remyelination in the adult central nervous system. eLife 8, e41869 (2019).

  66. Berghoff, S. A. et al. Dietary cholesterol promotes repair of demyelinated lesions in the adult brain. Nat. Commun. 8, 14241 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Yoon, H. et al. Interplay between exercise and dietary fat modulates myelinogenesis in the central nervous system. Biochim. Biophys. Acta 1862, 545–555 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Najm, F. J. et al. Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo. Nature 522, 216–220 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Mei, F. et al. Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat. Med. 20, 954–960 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Deshmukh, V. A. et al. A regenerative approach to the treatment of multiple sclerosis. Nature 502, 327–332 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Wang, S. et al. Anti-human TREM2 induces microglia proliferation and reduces pathology in an Alzheimer’s disease model. J Exp Med 217, e20200785 (2020).

  72. Berner, D. K. et al. Meprin beta cleaves TREM2 and controls its phagocytic activity on macrophages. FASEB J. 34, 6675–6687 (2020).

    Article  CAS  PubMed  Google Scholar 

  73. Sosic-Jurjevic, B. et al. The isoflavones genistein and daidzein increase hepatic concentration of thyroid hormones and affect cholesterol metabolism in middle-aged male rats. J. Steroid Biochem. Mol. Biol. 190, 1–10 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Thelen, K. M. et al. Brain cholesterol synthesis in mice is affected by high dose of simvastatin but not of pravastatin. J. Pharmacol. Exp. Ther. 316, 1146–1152 (2006).

    Article  CAS  PubMed  Google Scholar 

  75. Sampaio, J. L. et al. Membrane lipidome of an epithelial cell line. Proc. Natl Acad. Sci. USA 108, 1903–1907 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Ejsing, C. S. et al. Global analysis of the yeast lipidome by quantitative shotgun mass spectrometry. Proc. Natl Acad. Sci. USA 106, 2136–2141 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Herzog, R. et al. A novel informatics concept for high-throughput shotgun lipidomics based on the molecular fragmentation query language. Genome Biol. 12, R8 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Fitzner, D. et al. Cell-type- and brain-region-resolved mouse brain lipidome. Cell Rep 32, 108132 (2020).

    Article  CAS  PubMed  Google Scholar 

  79. Duewell, P. et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464, 1357–1361 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The work was supported by grants from the German Research Foundation (SPP2191, TRR128-2, TRR 274 Project ID 408885537, Koselleck Project HA1737/16-1, SyNergy Excellence Cluster, EXC2145, Projekt ID390857198), the Human Frontier Science Program (HFSP), the ERC (Consolidator Grant to M.S.), and the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation. M.B.Q. was supported by a Boehringer Ingelheim Fonds PhD fellowship. We would like to thank A. Rhomberg, G. Kislinger, A. Kerksiek and K. Karg for their technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

M.B.-Q. and M.S. conceived the project and designed experiments. M.B.-Q., L.C.-C., A.D., M. Schifferer., D.L., C.K. carried out experiments, K.S., L.V., I.A., T.M., M.P., L.M., G.D.P., K.M.M,, J.W.L, C.H developed and provided tools, M.B-Q., L.C-C., A.D., M. Simons, D.L., C.K. analysed the data or supervised data acquisition. M.B-Q. visualized the data, M.B-Q. and M.S wrote the manuscript, M.S supervised the project.

Corresponding author

Correspondence to Mikael Simons.

Ethics declarations

Competing interests

G.D.P., K.M.M and J.W.L are paid employees and shareholders of Denali Therapeutics Inc. C.K. is an employee of Lipotype.

Additional information

Peer review information Nature Metabolism thanks Michela Matteoli and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: George Caputa; Elena Bellafante.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 The plasma lipidome is altered by Western diet.

a, PCA analysis of CD and WD plasma samples. b, Mol% of the sample occupied by the different lipid classes in CD- and WD-fed mice (mean ± SD, two-tailed Welch’s t-test). c,e, Mol% of each sample with lipids containing a certain number of double bonds (c) or a certain acyl chain length (e) (mean ± SD, two-tailed Welch’s t-test). d, Heatmap showing the significantly (p value ≤ 0.01, fold ≥2.5) changed lipid species in plasma from CD- and WD-fed mice. Scale indicated on top of the heatmap. Increased values are indicated in orange, while decreased values are in blue. f, Quantification of the glucose level in blood at baseline (0 minutes) and 15, 30, 60 and 120 minutes after intraperitoneal glucose administration. (mean ± SD, two-tailed Welch’s t-test). g, PCA analysis of plasma from young (3-months old) versus old (12-months old) mice. h, Mol% of the sample occupied by the different lipid classes in plasma from young and old mice (mean ± SD, two-tailed Welch’s t-test). i,k, Mol% of each sample with lipids containing a certain number of double bonds (i) or a certain acyl chain length (k) in plasma from young and old mice (mean ± SD, two-tailed Welch’s t-test). j, Heatmap showing the significantly (p value ≤ 0.05, fold ≥1.3) changed lipid species in plasma from young and old mice. Scale indicated on top of the heatmap. Increased values are indicated in orange, while decreased values are in blue. P-values below 0.1 and n numbers are indicated in the figures. PCA: principal component analysis, CD: control diet, WD: Western diet, CE: cholesterol esters, Chol: cholesterol, DAG: diacylglycerol, LPC: lysophosphatidylcholine, PC: phosphatidylcholine, TAG: triacylglycerides, PI: Phosphatodylinositol. In d and j, the lipids are numbered according to the carbon length; number of double bonds; and number of hydroxyl groups (for example TAG 52;3;0).

Source data

Extended Data Fig. 2 Western diet feeding alters the brain lipidome of mice.

a, PCA analysis of white matter (WM) and grey matter (GM) samples from brain of WD- and CD-fed mice. b,c, Lipid classes changed in brain WM (b) and GM (c) between CD- and WD-fed mice (n = 3 for WD WM, CD GM and WD GM. N = 4 for CD WM, data represent mean ± SD, two-tailed Welch’s t-test). d,e, Heatmap showing the significantly (p value ≤ 0.5, fold ≥1.3) changed lipid species in the brain WM (d) and in the brain GM (e). Scale indicated on the right side of the heatmap. Increased values are indicated in orange, while decreased values are in blue. f,g, Mol% of each sample with lipids containing a certain number of double bonds (f) or a certain acyl chain length (g) (mean ± SD, two-tailed Welch’s t-test). The data is shown for those lipid species significantly (p value ≤ 0.05, fold ≥1.3) changed in the WM (n = 3 for WD WM, n = 4 for CD WM, data represent mean ± SD, two-tailed Welch’s t-test). h,i, Mol% of each sample with lipids containing a certain number of double bonds (h) or a certain acyl chain length (i). The data is shown for those lipid species significantly (p value ≤ 0.05, fold ≥1.3) changed in the GM (n = 3 for both groups, data represent mean ± SD, two-tailed Welch’s t-test). j,k, Mol% of the sample occupied by the different lipid classes in the brain WM (j) and GM (k) in CD- and WD-fed mice (mean ± SD, two-tailed Welch’s t-test). l, Quantification of the number of lipid droplets detected in the ventricle wall separating the corpus callosum from the lateral ventricles (solid lines indicate the mean, two-tailed Welch’s t-test). m, Representative image of a PLIN2 staining in the ventricle wall demonstrating the accumulation of lipid droplets in this structure. Scale bar: 5 µm. n, Images of the ventricle wall separating the corpus callosum from the second ventricle in CD- and WD-fed mice. Lipid droplets are labelled in pale green and marked with white arrows. Scale bar: 5 µm. o, Quantification of the percentage of IBA1+ cells containing PLIN2+ lipid droplets in the corpus callosum (solid lines indicate the mean, two-tailed Welch’s t-test). p, Example images of microglia containing PLIN2+ lipid droplets in their cytoplasm. On the top row, the raw image is displayed. On the bottom row, a 3D clip of the corresponding image is displayed, where the PLIN2+ signal inside the cell is indicated with a white arrow. Scale bar images left side: 3 µm, Scale bar images right side: 2 µm. q, Quantification of the percentage of IBA1+ area occupied by PLIN2+ signal (solid lines indicate the mean, two-tailed Welch’s t-test). r, Quantification of microglia density in the corpus callosum (solid lines indicate the mean, two-tailed Welch’s t-test). s, Measurement of IBA1+ signal intensity in the corpus callosum (two-tailed Welch’s t-test). t, Images of the corpus callosum (CC) and cortex (Ctx) stained with IBA1 to label microglia, GFAP to label astrocytes and PLIN2 to label lipid droplets. White arrows indicate lipid droplets within GFAP+ cells; blue arrows indicate lipid droplets within IBA1+ cells. Scale bar: 100 µm. u, Quantification of the percentage of GFAP+ area occupied by PLIN2+ signal (solid lines indicate the mean, two-tailed Welch’s t-test). v,w, Area (v) and intensity (w) of GFAP+ staining in the corpus callosum of CD- and WD-fed mice. P-values below 0.1 and n numbers are indicated in the figures; each dot represents one mouse. PCA: principal component analysis, CD: control diet, WD: Western diet, WM: white matter, GM: grey matter, LPC: lysophosphatidylcholine, PG: phosphatidylglycerol, PC: phosphatidylcholine, PE: phosphatidylethanolamine, PE O-: Phosphatidylethanolamine-ether, LPE: lysophosphatidylethanolamine, DAG: diacylglycerol, PC O-: Phosphatidylcholine-ether, PS: phosphatidylserine Ctx: cortex, CC: corpus callosum, PLIN2: Perilipin2. In d and e, the lipids are numbered according to the carbon length; number of double bonds; and number of hydroxyl groups (for example LPE 24;4;0).

Source data

Extended Data Fig. 3 Western diet consumption causes weak microglia activation.

a-c, Quantification of the percentage of CLEC7A+/IBA1+ (a), AXL+/IBA+ (b) and MAC2+/IBA1+ (c) cells over all the population of IBA1+ cells (solid lines indicate the mean, two-tailed Welch’s t-test). d, Quantification of the mean grey value of fluorescently labelled Evans Blue in whole brain sections of CD- and WD-fed mice (solid lines indicate the mean, two-tailed Welch’s t-test). e-g Images of microglia in the corpus callosum labelled with IBA1 and the activation markers CLEC7A (e), AXL (f) and MAC2 (g). White arrows indicate cells positive for both IBA1 and the corresponding activation marker. Scale bars: CLEC7A: 10 µm, AXL and MAC2: 20 µm. h, Images of two whole brain sections of CD- and WD-fed mice demonstrating the presence of fluorescently-labelled Evans Blue in the brain. Scale bar: 500 µm. P-values below 0.1 and n numbers are indicated in the figure; each dot represents one mouse. CD: control diet, WD: western diet.

Extended Data Fig. 4 The differential production of endogenous LXR ligands does not explain the poor induction of the LXR pathway observed in WD-fed mice.

a, Quantification of IBA1+ cell density in the demyelinated lesions at 4 and 7 dpi in CD- and WD-fed mice. b, Images demonstrating the density of IBA1+ cells in demyelinated lesions at 4 and 7 dpi. Scale bar: 20 µm. c, Quantification of the increase in APOE levels from 2 to 7 dpi in CD- and WD-fed mice (two-tail Welch’s t-test). d, Example images of the Western blots used for quantification of APOE levels. Molecular weights are indicated on the left side. APOE levels were normalized to α-TUBULIN (α-TUB). e,f, Quantification of the number of myelin- (e) and crystal- (f) loaded IBA1+ cells per mm2 in lesion of CD-, WD-, WD + FF- and WD + RG-fed mice. FF and RG treatments successfully promoted myelin clearance by phagocytes in WD-fed mice (one-way Brown-Forsythe and Welch ANOVA tests with multiple comparisons corrected by Dunnett T3 test). The references groups, CD and WD, are from Fig. 4e,f. g, Images of the demyelinated lesion in the corpus callosum at 14 dpi exemplifying myelin- and crystal-loaded IBA1+ cells in all treatment groups. Scale bar: 20 µm. h, Images demonstrating the co-localization of the cholesterol crystal signal obtained by reflection microscopy with either PLIN2 (green) or CD68 signal (magenta). Original images and 3D clips are shown. Scale bar: 2 µm. i, Diagram illustrating the production of the various natural liver X receptor (LXR) ligands measured in our assay. j-l, Quantification of the amounts of the endogenous LXR agonists desmosterol (j), 24-hydroxycholesterol (k) and 27-hydroxycholesterol (l) in the demyelinated lesions at 4 and 7 dpi in CD- and WD-fed mice. Each LXR ligand was normalized to the cholesterol amounts in the same lesion (two-way ANOVA followed by multiple comparisons correction with Sidak test). Solid lines in the graphs indicate the mean. P-values below 0.1 and n numbers are indicated in the figure; each dot represents one mouse. dpi: days post injection, CD: control diet, WD: Western diet, α-TUB: α-tubulin, Chol: cholesterol, FF: Fenofibrate, RG: rosiglitazone.

Source data

Extended Data Fig. 5 TGFβ signaling is increased in the unlesioned brain of WD-fed mice.

a,b, Overview images of MHCII+IBA1+ (a) and MAC2+IBA1+ (b) cells in the demyelinated lesions at 4 dpi. Scale bar: 20 µm. c, Images of the unlesioned corpus callosum of CD- and WD-fed mice demonstrating the accumulation of Tgfb1 and Tgfb2 particles visualized by RNA in situ hybridization. Scale bar: 20 µm. d, Fold change in Tgfb1 and Tgfb2 expression in the brain of WD-fed mice relative to CD-fed mice (two-tailed Welch’s t-test). e, Quantification of the number of RNA particles of Tgfb1 and Tgfb2 per nuclei in the corpus callosum in CD- and WD-fed mice (two-tailed Welch’s t-test). f, Images of the unlesioned corpus callosum of CD- and WD-fed mice demonstrating the Tgfb1 and Tgfb2 particles accumulating within GFAP&ALDH1L1+ cells in the unlesioned corpus callosum of CD- and WD-fed mice. Scale bar: 20 µm. g,h, Quantification of the number of Tgfb1 and Tgfb2 particles within IBA1+ (g) or GFAP&ALDH1L1+ cells (h) in the unlesioned corpus callosum of CD- and WD-fed mice (two-tailed Welch’s t-test). i, Quantification using ELISA of TGFβ1 in the 2 dpi lesions in CD and WD mice. No significant differences were observed (p = 0.14, two-tailed Welch’s t-test). j, Quantification of the pSMAD2+ area within IBA1+ area in the unlesioned corpus callosum (two-tailed Welch’s t-test). k, Images of the unlesioned CC of CD- and WD-fed mice demonstrating accumulation of pSMAD2+ signal within IBA1+ cells. White arrows point to IBA1+ cells accumulating low amounts of pSMAD2+ signal within their nucleus. Scale bar: 40 µm. Solid lines in the graphs indicate the mean. P-values below 0.1 and n numbers are indicated in the figure. dpi: days post injection, CD: control diet, WD: Western diet. CC: corpus callosum, Ctx: cortex.

Extended Data Fig. 6 Distribution of TGFβ particles in the demyelinated lesion.

a,b, Images of the fluorescent in situ hybridization of Tgfb1 and Tgfb2 in the demyelinated lesion at 2 dpi showing accumulation within IBA1+ (a) cells or within GFAP&ALDH1L1+ cells (b). Scale bar: 20 µm in the overview, 10 µm in the Zoom-in. c, Quantification of the percentage of Tgfb1 and Tgfb2 mRNA particles in IBA1+ or in GFAP&ALDH1L1+ cells over the total number of cells in a demyelinated lesion at 2 dpi. ((mean ± SD, two-tailed Welch’s t-test). d-f, Expression change in Tgfb1 and Tgfb2 induced by high glucose concentrations in cultured primary microglia (d), high lipid concentrations in cultured primary microglia (e) and high lipid concentrations in cultured primary astrocytes (f) (two-tailed paired t-test). Solid lines in the graphs indicate the mean. P-values below 0.1 and n numbers are indicated in the figure. dpi: days post injection, CD: control diet, WD: Western diet.

Extended Data Fig. 7 Blocking TGFβ signaling does not improve repair in old mice.

a, Fold change in Tgfbr2 expression in the demyelinated lesions of Tgfbr2 KO and control mice (two-tailed Welch’s t-test). b, Quantification of the number of Tgfbr2+ microglia in the unlesioned brains of Tgfbr2 KO and control mice (two-tailed Welch’s t test). c, Images of the unlesioned brain of Tgfbr2 KO and control mice demonstrating the accumulation of Tgfbr2 RNA particles within IBA1+ microglia. Scale bar: 20 µm. d, Quantification of the ratio of TGFβR2 signal over GAPDH signal in microglia isolated from Tgfbr2 KO and control mice (two-tailed t-test). e, Example images of the Western blots used for quantification of TGFβR2 levels of isolated microglia from Tgfbr2 KO and control mice. TGFβR2 levels were normalized to GAPDH. f,g, Quantification of the demyelination (f) and IBA1+ (g) volume at 4 dpi in Tgfbr2 KO and control mice fed WD. h,i, Quantification of the demyelination (h) and IBA1+ (i) volume at 4 dpi in Tgfbr2 control and KO mice fed CD. j, Quantification of Tgfb1 and Tgfb2 expression in the unlesioned brain of young (3-months old) and old (12-months old) mice by RT-qPCR (two-tailed Welch’s t-test). k,l, Quantification of demyelination k) and IBA1+ (l) volume at 14 dpi in young, old and old+GS mice (one-way Brown-Forsythe and Welch ANOVA tests with multiple comparisons corrected by Dunnett T3 test). m, Images of corpus callosum lesions of old and old+GS mice at 14 dpi. Scale bar: 200 µm. n,o, Quantification of the number of myelin- (n) and crystal- (o) loaded IBA1+ cells per mm2 of lesion (two-tailed Welch’s t-test). p, Images of the demyelinated lesion in the corpus callosum at 14 dpi exemplifying myelin- and crystal-loaded IBA1+ cells in old and old+GS mice. Scale bar: 20 µm. N numbers are indicated in the figure; each dot represents one mouse. Solid lines in the graphs indicate the mean. CD: control diet, WD: Western diet GS: Galunisertib, dpi: days post injection.

Source data

Extended Data Fig. 8 4D9 treatment prevents myelin-induced TREM2 shedding.

a,d, Quantification using ELISA of cellular TREM2 in the unlesioned brain of old (12-months old) and young (3-months old) mice (a) and of CD- and WD-fed mice (d) (two-tailed Welch’s t-test). b,e, Quantification using ELISA of soluble TREM2 in the unlesioned brain old and young mice (b) and of CD- and WD-fed mice (e) (two-tailed Welch’s t-test). c, Change in expression of Trem2 in the demyelinated lesions of old mice at 2 dpi when compared to young mice (two-tailed Welch’s t-test). f,g, Quantification of the band density in Western blots of TREM2-labelled (f) and sTREM2-labelled (g) bands normalized by the density of the bands labelled by α-TUBULIN (α-TUB). Microglia were treated with myelin for 24 hours (one-way Brown-Forsythe and Welch ANOVA tests with multiple comparisons corrected by Dunnett T3 test). h, Example images of the Western blots used for quantification of TREM2 and sTREM2 densities. Molecular weight ladders are indicated on the left side. i, Quantification by ELISA of the amounts of sTREM2 found in the media in different conditions. Microglia were treated with 20 µg/mL of 4D9 antibody or IgG isotype control for 16 hours, followed by 24 hours of myelin treatment (two-tailed Welch’s t-test). P-values and n numbers are indicated in the figure. Solid lines in the graphs indicate the mean. dpi: days post injection, CD: control diet, WD: Western diet, sTREM2: soluble TREM2, Mye5: 5 µg/mL myelin treatment, Mye8: 8 µg/mL myelin treatment, Mye30: 30 µg/mL myelin treatment, 4D9: TREM2 enhancing antibody.

Source data

Supplementary information

Source data

Source Data Extended Data Fig. 1

Source data for lipidomics analysis

Source Data Extended Data Fig. 2

Source data for lipidomics analysis

Source Data Extended Data Fig. 4

Unprocessed Western Blots for Extended Data Fig. 4d

Source Data Extended Data Fig. 7

Unprocessed Western Blots for Extended Data Fig. 7e

Source Data Extended Data Fig. 8

Unprocessed Western Blots for Extended Data Fig. 8h

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

Bosch-Queralt, M., Cantuti-Castelvetri, L., Damkou, A. et al. Diet-dependent regulation of TGFβ impairs reparative innate immune responses after demyelination. Nat Metab 3, 211–227 (2021). https://doi.org/10.1038/s42255-021-00341-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00341-7

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