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:

A class of anti-inflammatory lipids decrease with aging in the central nervous system

Abstract

Lipids contribute to the structure, development, and function of healthy brains. Dysregulated lipid metabolism is linked to aging and diseased brains. However, our understanding of lipid metabolism in aging brains remains limited. Here we examined the brain lipidome of mice across their lifespan using untargeted lipidomics. Co-expression network analysis highlighted a progressive decrease in 3-sulfogalactosyl diacylglycerols (SGDGs) and SGDG pathway members, including the potential degradation products lyso-SGDGs. SGDGs show an age-related decline specifically in the central nervous system and are associated with myelination. We also found that an SGDG dramatically suppresses LPS-induced gene expression and release of pro-inflammatory cytokines from macrophages and microglia by acting on the NF-κB pathway. The detection of SGDGs in human and macaque brains establishes their evolutionary conservation. This work enhances interest in SGDGs regarding their roles in aging and inflammatory diseases and highlights the complexity of the brain lipidome and potential biological functions in aging.

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: Global lipidomics of mouse brain during aging.
Fig. 2: Discovery of a class of lipids that progressively decrease during aging.
Fig. 3: Quantification of SGDGs in mouse tissues.
Fig. 4: SGDGs are present in human.
Fig. 5: Lyso-SGDGs are the potential degradation products of SGDGs in the brain.
Fig. 6: SGDG exhibits anti-inflammatory effects via the NF-κB pathway.

Similar content being viewed by others

Data availability

The lipidomics raw data and metadata have been deposited into MassIVE public repository with the accession number MSV000090179 (https://doi.org/10.25345/C59Z90G8H). LipidBlast, Lipid MAPS, and METLIN are available at https://fiehnlab.ucdavis.edu/projects/lipidblast, https://www.lipidmaps.org/, and https://metlin.scripps.edu/landing_page.php?pgcontent=mainPage, respectively. Source data are provided with this paper.

References

  1. Mattson, M. P. & Arumugam, T. V. Hallmarks of brain aging: adaptive and pathological modification by metabolic states. Cell Metab. 27, 1176–1199 (2018).

    Article  CAS  Google Scholar 

  2. Bishop, N. A., Lu, T. & Yankner, B. A. Neural mechanisms of ageing and cognitive decline. Nature 464, 529–535 (2010).

    Article  CAS  Google Scholar 

  3. Wenk, M. R. The emerging field of lipidomics. Nat. Rev. Drug Discov. 4, 594–610 (2005).

    Article  CAS  Google Scholar 

  4. Hallett, P. J., Engelender, S. & Isacson, O. Lipid and immune abnormalities causing age-dependent neurodegeneration and Parkinson’s disease. J. Neuroinflammation 16, 1–15 (2019).

    Article  CAS  Google Scholar 

  5. Bu, G. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat. Rev. Neurosci. 10, 333–344 (2009).

    Article  CAS  Google Scholar 

  6. Lwin, A., Orvisky, E., Goker-Alpan, O., LaMarca, M. E. & Sidransky, E. Glucocerebrosidase mutations in subjects with parkinsonism. Mol. Genet. Metab. 81, 70–73 (2004).

    Article  CAS  Google Scholar 

  7. Blankman, J. L., Long, J. Z., Trauger, S. A., Siuzdak, G. & Cravatt, B. F. ABHD12 controls brain lysophosphatidylserine pathways that are deregulated in a murine model of the neurodegenerative disease PHARC. Proc. Natl Acad. Sci. 110, 1500–1505 (2013).

    Article  CAS  Google Scholar 

  8. Li, Q. et al. Changes in lipidome composition during brain development in humans, chimpanzees, and macaque monkeys. Mol. Biol. evolution 34, 1155–1166 (2017).

    Article  Google Scholar 

  9. Yu, Q. et al. Lipidome alterations in human prefrontal cortex during development, aging, and cognitive disorders. Mol. Psychiatry 25, 2952–2969 (2020).

    Article  CAS  Google Scholar 

  10. Tu, J., Yin, Y., Xu, M., Wang, R. & Zhu, Z.-J. Absolute quantitative lipidomics reveals lipidome-wide alterations in aging brain. Metabolomics 14, 1–11 (2018).

    Article  CAS  Google Scholar 

  11. Ding, J. et al. A metabolome atlas of the aging mouse brain. Nat. Commun. 12, 1–12 (2021).

    Article  Google Scholar 

  12. Saghatelian, A. et al. Assignment of endogenous substrates to enzymes by global metabolite profiling. Biochemistry 43, 14332–14339 (2004).

    Article  CAS  Google Scholar 

  13. Yore, M. M. et al. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell 159, 318–332 (2014).

    Article  CAS  Google Scholar 

  14. Amor, S., Puentes, F., Baker, D. & Van Der Valk, P. Inflammation in neurodegenerative diseases. Immunology 129, 154–169 (2010).

    Article  CAS  Google Scholar 

  15. Wyss-Coray, T. & Mucke, L. Inflammation in neurodegenerative disease—a double-edged sword. Neuron 35, 419–432 (2002).

    Article  CAS  Google Scholar 

  16. Sindelar, M. & Patti, G. J. Chemical discovery in the era of metabolomics. JACS 142, 9097–9105 (2020).

    Article  CAS  Google Scholar 

  17. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinf. 9, 1–13 (2008).

    Article  Google Scholar 

  18. Cutler, R. G. et al. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc. Natl Acad. Sci. 101, 2070–2075 (2004).

    Article  CAS  Google Scholar 

  19. Li, M.-L. et al. 547 transcriptomes from 44 brain areas reveal features of the aging brain in non-human primates. Genome Biol. 20, 1–17 (2019).

    Article  Google Scholar 

  20. Horvath, S. et al. Analysis of oncogenic signaling networks in glioblastoma identifies ASPM as a molecular target. Proc. Natl Acad. Sci. 103, 17402–17407 (2006).

    Article  CAS  Google Scholar 

  21. Toida, T., Chaidedgumjorn, A. & Linhardt, R. J. Structure and bioactivity of sulfated polysaccharides. Trends Glycosci. Glycotechnol. 15, 29–46 (2003).

    Article  CAS  Google Scholar 

  22. Gorelik, D., Lin, Y. C., Briceno-Strocchia, A. I. & Taylor, M. S. Diarylborinic acid-catalyzed, site-selective sulfation of carbohydrate derivatives. J. Org. Chem. 84, 900–908 (2019).

    Article  CAS  Google Scholar 

  23. Kind, T. et al. LipidBlast in silico tandem mass spectrometry database for lipid identification. Nat. Methods 10, 755–758 (2013).

    Article  CAS  Google Scholar 

  24. Sud, M. et al. Lmsd: lipid maps structure database. Nucleic Acids Res. 35, D527–D532 (2007).

    Article  CAS  Google Scholar 

  25. Smith, C. A. et al. METLIN: a metabolite mass spectral database. Therapeutic drug Monit. 27, 747–751 (2005).

    Article  CAS  Google Scholar 

  26. Flynn, T. J., Deshmukh, D., Rao, G. S. & Pieringer, R. A. Sulfogalactosyl diacylglycerol: occurrence and biosynthesis of a novel lipid in rat brain. Biochem. Biophys. Res. Commun. 65, 122–128 (1975).

    Article  CAS  Google Scholar 

  27. Ishizuka, I., Inomata, M., Ueno, K. & Yamakawa, T. Sulfated glyceroglycolipids in rat brain. Structure sulfation in vivo, and accumulation in whole brain during development. J. Biol. Chem. 253, 898–907 (1978).

    Article  CAS  Google Scholar 

  28. Ishizuka, I. & Inomata, M. Sulphated glycoglycerolipids in rat brain: decrease and disappearance after developmental age. J. Neurochem. 33, 387–388 (1979).

    Article  CAS  Google Scholar 

  29. Baillie, T.A. (ed.) Stable Isotopes: Applications in Pharmacology, Toxicology and Clinical Research. (Palgrave Macmillan, 1978).

  30. Willott, J. F., Bross, L. S. & McFadden, S. L. Morphology of the dorsal cochlear nucleus in C57BL/6J and CBA/J mice across the life span. J. Comp. Neurol. 321, 666–678 (1992).

    Article  CAS  Google Scholar 

  31. Ishizuka, I. Chemistry and functional distribution of sulfoglycolipids. Prog. Lipid Res. 36, 245–319 (1997).

    Article  CAS  Google Scholar 

  32. Pieringer, J., Rao, G. S., Mandel, P. & Pieringer, R. A. The association of the sulphogalactosylglycerolipid of rat brain with myelination. Biochem. J. 166, 421–428 (1977).

    Article  CAS  Google Scholar 

  33. Chernoff, G. F. Shiverer: an autosomal recessive mutant mouse with myelin deficiency. J. Heredity 72, 128–128 (1981).

    Article  CAS  Google Scholar 

  34. Rao, G. S., Norcia, L. N., Pieringer, J. & Pieringer, R. A. The biosynthesis of sulphogalactosyldiacylglycerol of rat brain in vitro. Biochem. J. 166, 429–435 (1977).

    Article  CAS  Google Scholar 

  35. Erwig, M. S. et al. Myelin: Methods for Purification and Proteome Analysis. Methods Mol. Biol. 1936, 37–63 (2019).

    Article  CAS  Google Scholar 

  36. Gopalakrishnan, G. et al. Lipidome and proteome map of myelin membranes. J. Neurosci. Res. 91, 321–334 (2013).

    Article  CAS  Google Scholar 

  37. Honke, K. Biosynthesis and biological function of sulfoglycolipids. Proc. Jpn. Acad., Ser. B 89, 129–138 (2013).

    Article  CAS  Google Scholar 

  38. Tanphaichitr, N. et al. Properties, metabolism and roles of sulfogalactosylglycerolipid in male reproduction. Prog. Lipid Res. 72, 18–41 (2018).

    Article  CAS  Google Scholar 

  39. McKhann, G. M. & Ho, W. The in vivo and in vitro synthesis of sulphatides during development. J. Neurochem. 14, 717–724 (1967).

    Article  CAS  Google Scholar 

  40. Burkart, T., Caimi, L., Herschkowitz, N. N. & Wiesmann, U. N. Metabolism of sulfogalactosyl glycerolipids in the myelinating mouse brain. Dev. Biol. 98, 182–186 (1983).

    Article  CAS  Google Scholar 

  41. Wang, C., Wang, J., Qin, C. & Han, X. Analysis of monohexosyl alkyl (alkenyl)-acyl glycerol in brain samples by shotgun lipidomics. Anal. Chim. Acta 1129, 143–149 (2020).

    Article  CAS  Google Scholar 

  42. Lee, C.-K., Weindruch, R. & Prolla, T. A. Gene-expression profile of the ageing brain in mice. Nat. Genet. 25, 294–297 (2000).

    Article  CAS  Google Scholar 

  43. Hannun, Y. A. & Obeid, L. M. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat. Rev. Mol. Cell Biol. 9, 139–150 (2008).

    Article  CAS  Google Scholar 

  44. Guijas, C., Montenegro-Burke, J. R., Warth, B., Spilker, M. E. & Siuzdak, G. Metabolomics activity screening for identifying metabolites that modulate phenotype. Nat. Biotechnol. 36, 316–320 (2018).

    Article  CAS  Google Scholar 

  45. Chiurchiù, V., Leuti, A. & Maccarrone, M. Bioactive lipids and chronic inflammation: managing the fire within. Front. Immunol. 9, 38 (2018).

    Article  Google Scholar 

  46. Franceschi, C. & Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. Ser. A Biomed. Sci. Med. Sci. 69, S4–S9 (2014).

    Article  Google Scholar 

  47. Franceschi, C. et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev. 128, 92–105 (2007).

    Article  CAS  Google Scholar 

  48. Ye, S.-M. & Johnson, R. W. Increased interleukin-6 expression by microglia from brain of aged mice. J. Neuroimmunol. 93, 139–148 (1999).

    Article  CAS  Google Scholar 

  49. Porcher, L. et al. Aging triggers an upregulation of a multitude of cytokines in the male and especially the female rodent hippocampus but more discrete changes in other brain regions. J. Neuroinflammation 18, 219 (2021).

    Article  CAS  Google Scholar 

  50. Peters, A. The effects of normal aging on myelin and nerve fibers: a review. J. Neurocytol. 31, 581–593 (2002).

    Article  Google Scholar 

  51. Turturro, A. et al. Growth curves and survival characteristics of the animals used in the biomarkers of aging program. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 54, B492–B501 (1999).

    Article  CAS  Google Scholar 

  52. Liebisch, G. et al. Shorthand notation for lipid structures derived from mass spectrometry. J. Lipid Res. 54, 1523–1530 (2013).

    Article  CAS  Google Scholar 

  53. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Physiol. 37, 911–917 (1959).

    Article  CAS  Google Scholar 

  54. Tautenhahn, R., Patti, G. J., Rinehart, D. & Siuzdak, G. XCMS Online: a web-based platform to process untargeted metabolomic data. Anal. Chem. 84, 5035–5039 (2012).

    Article  CAS  Google Scholar 

  55. Kessner, D., Chambers, M., Burke, R., Agus, D. & Mallick, P. ProteoWizard: open source software for rapid proteomics tools development. Bioinformatics 24, 2534–2536 (2008).

    Article  CAS  Google Scholar 

  56. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  Google Scholar 

  57. Ip, W. E., Hoshi, N., Shouval, D. S., Snapper, S. & Medzhitov, R. Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science 356, 513–519 (2017).

    Article  CAS  Google Scholar 

  58. Moraes‐Vieira, P. M. et al. Leptin deficiency impairs maturation of dendritic cells and enhances induction of regulatory T and Th17 cells. Eur. J. Immunol. 44, 794–806 (2014).

    Article  Google Scholar 

  59. Moraes-Vieira, P. M. et al. RBP4 activates antigen-presenting cells, leading to adipose tissue inflammation and systemic insulin resistance. Cell Metab. 19, 512–526 (2014).

    Article  CAS  Google Scholar 

  60. Dugas, J. C. & Emery, B. Purification of oligodendrocyte precursor cells from rat cortices by immunopanning. Cold Spring Harb. Protoc. 2013, 745–758 (2013).

    Article  Google Scholar 

Download references

Acknowledgements

We thank members of the Saghatelian laboratory for helpful advice and discussions; R. Rissman for providing the human hippocampus sample; and J. Ning for providing the anti-PLP antibody. This research was supported by Ferring Pharmaceuticals and Frederik Paulsen (D.S. and A.S.), NIH (P30 CA014195, R01DK106210, A.S.), the Wu Tsai Human Performance Alliance (A.S), ONPRC (P51 OD 010092, S.G.K.), the Anderson Foundation (D.T.), the Bruce Ford and Anne Smith Bundy Foundation (D.T.), Pioneer Fellowship (D.T.), NIH (R01NS119823, J.B.Z.), the Howard Hughes Medical Institute (A.D.), CZI Neurodegeneration Network (N.J.A.), FAPESP (2017/01184-9, M.A.M.), and NIH (R01AG069206, RF1AG061296, P.M.)

Author information

Authors and Affiliations

Authors

Contributions

D.T. and A.S. conceived the project, designed the experiments, and analyzed and interpreted the data. D.T. performed the lipidomics, LC–MS/MS, biochemical, and cell-based experiments. S.K. synthesized lipid standards. M.E.E. collected mouse tissues. P.Z. performed the co-expression network analysis. J.W. extracted lipids and collected mouse tissues. T.C. performed cell-based experiments. A.F.M.P. contributed to LC–MS/MS analysis. A.R. prepared BMDMs. C.J.D. and J.M.V. performed the NF-κB luciferase assay. R.G.L. and M.A.M. provided the calorie restriction samples. E.W. and A.D. provided the 29-month-old and 5-month-old mouse tissues. M.I. and J.B.Z. provided OPCs and differentiated oligodendrocytes. P.M. contributed to the BV2 experiments. N.J.A. contributed to the shiverer mouse experiments. S.G.K. provided macaque tissues. D.T., S.K., D.S., and A.S. wrote the manuscript. P.C.G., P.Z., M.E.E., P.M., and S.G.K. edited the manuscript. A.S. and D.S. supervised the project.

Corresponding authors

Correspondence to Dionicio Siegel or Alan Saghatelian.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Huiyong Yin, Reinald Pamplona and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Average levels of the co-expressed lipids in the 23 modules across different ages.

n = 4 mice/group. Values are expressed with box plots. Boxes represent 25th to 75th percentile. Center lines represent the median values. Whiskers represent 1.5× interquartile range.

Extended Data Fig. 2 Association between module eigenlipid and age.

Blue line represents fitted linear regression line between module eigenlipid and age. Pink shaded area represents 95% confidence interval of the regression line. Modules that are significantly associated with age are highlighted with red boxes.

Extended Data Fig. 3 Quantification of SGDGs in the brain of mice with three months of calorie restriction.

Sex, male; age, 6 months. n = 8 mice/group, data are means ± SEM. *P < 0.05 versus ad libitum-fed mice. P values were 0.0217, 0.0155, 0.0276, 0.0453, 0.0410, 0.0243, 0.2387, 0.0694, 0.1417, 0.1103, 0.0423, 0.0451 (left to right). Statistical significance was calculated using unpaired, two-tailed Student’s t-test with equal variance.

Extended Data Fig. 4 Fractionation of the mouse brain lysate into membrane and soluble fractions revealed that SGDGs are largely membrane-associated.

Bar graph shows SGDG levels relative to the total lysate. n = 3, data are means ± SEM.

Extended Data Fig. 5 Quantification of SGAAGs in mouse tissues.

(a) Quantification of SGAAGs in the brain and spinal cord of WT female mice at different ages. n = 4 mice/group, data are means ± SEM. (b) Quantification of SGAAGs in the brain, spinal cord, and testis of WT male mice at different ages. Data were collected from six 3-month-old, three 7-month-old, and six 22-month-old mice. Data are means ± SEM.

Extended Data Fig. 6 SGDGs are enriched in myelin.

(a) Quantification of sulfatides in purified mouse brain myelin. n = 3 mice, data are means ± SEM. (b) Validation of myelin purification. Immunoblot shows that myelin proteins, PLP and MBP, are enriched in purified rat brain myelin compared with the brain lysate, whereas the abundant neuronal protein synaptophysin is depleted. (c) Quantification of SGDGs and sulfatides in purified rat brain myelin. n = 2 technical replicates, data are means ± SEM.

Source data

Extended Data Fig. 7

Quantification of SGDGs in oligodendrocyte precursor cells and myelinated oligodendrocytes at day 5 of differentiation.

Extended Data Fig. 8 Quantification of SGDGs in macaque and human brains.

(a) Extracted ion chromatograms of SGDG(14:0_16:0) from the parietal cortex of a 6-year-old male macaque and the internal standard SGDG(13C16-16:0/14:0). (b) Quantification of SGDGs in the parietal cortex of a 6-year-old male macaque. (c) Quantification of SGDGs in a frontal brain sample collected postmortem from a 5-month-old male subject.

Extended Data Fig. 9

Expression profile of MGDGs, SGAAGs, SGDGs, MGMGs, and lyso-SGDGs in the spinal cord module which shows progressive decrease during aging.

Extended Data Fig. 10 SGDG exhibits anti-inflammatory effects in RAW 264.7 cells and BV2 cells.

(a) RAW 264.7 cells were incubated with media alone, 100 ng/mL of LPS, or co-treated with 100 ng/mL of LPS and 5 μM of SGDG(14:0/16:0) for 4 hours. mRNA levels were determined by RT-PCR. n = 4 replicates. P values were 3.24e-13, 9.54e-11, 3.44e-9, 3.22e-7, 9.35e-11, 5.41e-7, 1.36e-10, 0.00004, 7.10e-10, 0.00001, 2.17e-6, 0.06078 (left to right, top to bottom). (b) BV2 cells were incubated with media alone, 100 ng/mL of LPS, or co-treated with 100 ng/mL of LPS and 25 μM of SGDG(14:0/16:0) or dexamethasone (Dex) for 24 hours. IL-6 and TNF-α levels in the media were measured by ELISA and normalized to cell viability. n = 3 replicates. P values were 0, 0, 0, 0, 0.00005, 0.00038 (left to right). In a, b, data are means ± SEM. *P < 0.05 versus LPS. Statistical significance was calculated using one-way ANOVA followed by post hoc Dunnett’s test to correct for multiple comparisons.

Supplementary information

Supplementary Information

Supplementary Figs. 1–7, Supplementary Table 1, and Supplementary Note.

Reporting Summary

Source data

Source Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor 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

Tan, D., Konduri, S., Erikci Ertunc, M. et al. A class of anti-inflammatory lipids decrease with aging in the central nervous system. Nat Chem Biol 19, 187–197 (2023). https://doi.org/10.1038/s41589-022-01165-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01165-6

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