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:

Neuronal accumulation of peroxidated lipids promotes demyelination and neurodegeneration through the activation of the microglial NLRP3 inflammasome

Abstract

Peroxidated lipids accumulate in the presence of reactive oxygen species and are linked to neurodegenerative diseases. Here we find that neuronal ablation of ARF1, a small GTPase important for lipid homeostasis, promoted accumulation of peroxidated lipids, lipid droplets and ATP in the mouse brain and led to neuroinflammation, demyelination and neurodegeneration, mainly in the spinal cord and hindbrain. Ablation of ARF1 in cultured primary neurons led to an increase in peroxidated lipids in co-cultured microglia, activation of the microglial NLRP3 inflammasome and release of inflammatory cytokines in an Apolipoprotein E-dependent manner. Deleting the Nlrp3 gene rescued the neurodegenerative phenotypes in the neuronal Arf1-ablated mice. We also observed a reduction in ARF1 in human brain tissue from patients with amyotrophic lateral sclerosis and multiple sclerosis. Together, our results uncover a previously unrecognized role of peroxidated lipids released from damaged neurons in activation of a neurotoxic microglial NLRP3 pathway that may play a role in human neurodegeneration.

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: ARF1 ablation promotes demyelination, axon degeneration, synapse loss and fragmentation of neurofilament.
Fig. 2: Neuron-specific ablation of ARF1 induces the neurodegenerative phenotypes in mice.
Fig. 3: ARF1-KO neurons release peroxidated lipids.
Fig. 4: Peroxidated lipids promote neurodegeneration through the NLRP3 inflammasome.
Fig. 5: Neurodegeneration phenotypes of neuronal ARF1-KO mice were suppressed by an NLRP3 inhibitor or in Nlrp3−/− mice.
Fig. 6: An ARF1-reduction-associated neuroinflammation pathway exists in patient with MS and ALS.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited in the Gene Expression Omnibus under accession no. GSE183483. All other data supporting the findings of this study are available within the source data provided with this paper and its Supplementary Information and are available from the corresponding author upon reasonable request.

Code availability

There is no new code was created for analysis of the RNA-seq data.

References

  1. Negre-Salvayre, A., Coatrieux, C., Ingueneau, C. & Salvayre, R. Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br. J. Pharmacol. 153, 6–20 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Petrovic, S., Arsic, A., Ristic-Medic, D., Cvetkovic, Z. & Vucic, V. Lipid peroxidation and antioxidant supplementation in neurodegenerative diseases: a review of human studies. Antioxidants 9, 1128 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  3. Bailey, A. P. et al. Antioxidant role for lipid droplets in a stem cell niche of Drosophila. Cell 163, 40–53 (2015).

    Article  Google Scholar 

  4. Yang, W. S. et al. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl Acad. Sci. USA 113, E4966–E4975 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Beal, M. F. et al. Isotope-reinforced polyunsaturated fatty acids improve Parkinson’s disease-like phenotype in rats overexpressing α-synuclein. Acta Neuropathol. Commun. 8, 220 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nowak, J. Z. Oxidative stress, polyunsaturated fatty acids-derived oxidation products and bisretinoids as potential inducers of CNS diseases: focus on age-related macular degeneration. Pharmacol. Rep. 65, 288–304 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Mao, X. Y., Zhou, H. H. & Jin, W. L. Redox-related neuronal death and crosstalk as drug targets: focus on epilepsy. Front. Neurosci. 13, 512 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Ioannou, M. S. et al. Neuron-astrocyte metabolic coupling protects against activity-induced fatty acid toxicity. Cell 177, 1522–1535 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Liu, L. et al. Glial lipid droplets and ROS induced by mitochondrial defects promotes neurodegeneration. Cell 160, 177–190 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ackema, K. B. et al. The small GTPase Arf1 modulates mitochondrial morphology and function. EMBO J. 33, 2659–2675 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kaczmarek, B., Verbavatz, J. M. & Jackson, C. L. GBF1 and Arf1 function in vesicular trafficking, lipid homoeostasis and organelle dynamics. Biol. Cell 109, 391–399 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Nagashima, S. et al. Golgi-derived PI (4) P-containing vesicles drive late steps of mitochondrial division. Science 367, 1366–1371 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Wilfling, F. et al. Arf1/COPI machinery acts directly on lipid droplets and enables their connection to the ER for protein targeting. eLife 3, e01607 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Liu, L. et al. The glia–neuron lactate shuttle and elevated ROS promote lipid synthesis in neurons and lipid droplet accumulation in glia via APOE/D. Cell Metab. 26, 719–737 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Xiang, Y. et al. Active ADP-ribosylation factor-1 (ARF1) is required for mitotic Golgi fragmentation. J. Biol. Chem. 282, 21829–21837 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Boulay, P. L., Cotton, M., Melançon, P. & Claing, A. ADP-ribosylation factor 1 controls the activation of the phosphatidylinositol 3-kinase pathway to regulate epidermal growth factor-dependent growth and migration of breast cancer cells. J. Biol. Chem. 283, 36425–36434 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wonderlich, E. R. et al. ADP-ribosylation factor 1 activity is required to recruit AP-1 to the major histocompatibility complex class I (MHC-I) cytoplasmic tail and disrupt MHC-I trafficking in HIV-1-infected primary T cells. J. Virol. 85, 12216–12226 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sumiyoshi, M. et al. Arf1 and Arf6 synergistically maintain survival of T cells during activation. J. Immunol. 206, 366–375 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Roy, N. S. et al. Interaction of the N terminus of ADP-ribosylation factor with the PH domain of the GTPase-activating protein ASAP1 requires phosphatidylinositol 4,5-bisphosphate. J. Biol. Chem. 294, 17354–17370 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lopes-da-Silva, M. et al. A GBF1-dependent mechanism for environmentally responsive regulation of ER-Golgi transport. Dev. Cell 49, 786–801 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Singh, S. R. et al. The lipolysis pathway sustains normal and transformed stem cells in adult Drosophila. Nature 538, 109–113 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, G. et al. Arf1-mediated lipid metabolism sustains cancer cells and its ablation induces anti-tumor immune responses in mice. Nat. Commun. 11, 220 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Miyamoto, Y. et al. BIG1/Arfgef1 and Arf1 regulate the initiation of myelination by Schwann cells in mice. Sci. Adv. 4, eaar4471 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Liu, Y., Wang, Y., Ding, W. & Wang, Y. Mito-TEMPO alleviates renal fibrosis by reducing inflammation, mitochondrial dysfunction, and endoplasmic reticulum stress. Oxid. Med. Cell Longev. 2018, 5828120 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kang, R. et al. Lipid peroxidation drives gasdermin D-mediated pyroptosis in lethal polymicrobial sepsis. Cell Host Microbe. 24, 97–108 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen, R. et al. cAMP metabolism controls caspase-11 inflammasome activation and pyroptosis in sepsis. Sci. Adv. 5, eaav5562 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Coll, R. C. et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat. Med. 21, 2482–2455 (2015).

    Article  Google Scholar 

  28. Jiang, X., Stockwell, B. R. & Conrad, M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Lin, M. T. & Beal, M. F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443, 787–795 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Gandhi, S. & Abramov, A. Y. Mechanism of oxidative stress in neuro-degeneration. Oxid. Med. Cell Longev. 2012, 428010 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Gilgun-Sherki, Y., Melamed, E. & Offen, D. Oxidative stress induced-neurodegenerative diseases: the need for antioxidants that penetrate the blood–brain barrier. Neuropharmacology 40, 959–975 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Ising, C. et al. NLRP3 inflammasome activation drives tau pathology. Nature 575, 669–673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Scheiblich, H., Trombly, M., Ramirez, A. & Heneka, M. T. Neuroimmune connections in aging and neurodegenerative diseases. Trends Immunol. 41, 300–312 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Venegas, C. et al. Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer’s disease. Nature 552, 355–361 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Galasko, D. R. et al. Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol. 69, 836–841 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Jassim, A. H., Inman, D. M. & Mitchell, C. H. Crosstalk between dysfunctional mitochondria and inflammation in glaucomatous neurodegeneration. Front. Pharmacol. 12, 699623 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Solini, A. et al. P2X7 receptor/NLRP3 inflammasome complex and α-synuclein in peripheral blood mononuclear cells: a prospective study in neo-diagnosed, treatment-naive Parkinson’s disease. Eur. J. Neurol. 28, 2648–2656 (2021).

    Article  PubMed  Google Scholar 

  38. Piancone, F. et al. Inflammatory responses to monomeric and aggregated α-synuclein in peripheral blood of parkinson disease patients. Front. Neurosci. 15, 639646 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Trudler, D. et al. Soluble α-synuclein-antibody complexes activate the NLRP3 inflammasome in hiPSC-derived microglia. Proc. Natl Acad. Sci. USA 118, e2025847118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hummel, C. et al. Expression and cell type-specific localization of inflammasome sensors in the spinal cord of SOD1 (G93A) mice and sporadic amyotrophic lateral sclerosis patients. Neuroscience 463, 288–302 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Deora, V. et al. The microglial NLRP3 inflammasome is activated by amyotrophic lateral sclerosis proteins. Glia 68, 407–421 (2020).

    Article  PubMed  Google Scholar 

  42. Lehmann, S. et al. Expression profile of pattern recognition receptors in skeletal muscle of SOD1 (G93A) amyotrophic lateral sclerosis (ALS) mice and sporadic ALS patients. Neuropathol. Appl. Neurobiol. 44, 606–627 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Kadowaki, A. & Quintana, F. J. The NLRP3 inflammasome in progressive multiple sclerosis. Brain 143, 1286–1288 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lemprière, S. NLRP3 inflammasome activity as biomarker for primary progressive multiple sclerosis. Nat. Rev. Neurol. 16, 350 (2020).

    Article  PubMed  Google Scholar 

  45. Siew, J. J. et al. Galectin-3 is required for the microglia-mediated brain inflammation in a model of Huntington’s disease. Nat. Commun. 10, 3473 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Paldino, E., D’Angelo, V., Sancesario, G. & Fusco, F. R. Pyroptotic cell death in the R6/2 mouse model of Huntington’s disease: new insight on the inflammasome. Cell Death Discov. 6, 69 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang, J. et al. Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency. Nature 588, 459–465 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hong, S. et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ge, X. et al. Missense-depleted regions in population exomes implicate Ras superfamily nucleotide-binding protein alteration in patients with brain malformation. NPJ Genom. Med. 1, 16036 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Han, W. et al. Shisa7 is a GABA A receptor auxiliary subunit controlling benzodiazepine actions. Science 366, 246–250 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lian, H., Roy, E. & Zheng, H. Protocol for primary microglial culture preparation. Bio. Protoc. 6, e1989 (2016).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank the Pathology/Histotechnology Laboratory at the Frederick National Laboratory for Cancer Research for help with tissue sectioning and K. Nagashima at the Electron Microscopy Laboratory of the NCI, NIH for help with EM experiments. We thank A. Abdelmaksoud, U. Sehgal, M. Cam, P. Jailwala, Y. Zhao, T.W. Shen and S. Kuhn at NCI, NIH for help with analyzing RNA-seq data; D. Reich at the NINDS for help with getting human samples; J.M. Wang at NCI for help supervising the protocol for mice research; S. Lopez at NCI for help editing the manuscript. We thank the NIH NeuroBioBank and the Rocky Mountain Multiple Sclerosis Center Tissue Bank for providing healthy, ALS and MS human samples. This research was supported by the Intramural Research Program of the NIH, NCI and Center for Cancer Research (S.X.H.); NINDS intramural program (W.L.); the National Natural Science Foundation of China (92057205 to S.X.H.). NCI and NINDS at NIH supported the study design, purchase of reagents, data collection and analysis, revision and publishing process and preparation of the manuscript. The National Natural Science Foundation of China supported the publishing process.

Author information

Authors and Affiliations

Authors

Contributions

G.W. and S.X.H. conceived and designed the experiments. G.W. and W.Y. performed the majority of experiments. H.S. and Q.T. assisted with experiments. G.W. and S.X.H. analyzed data. S.X.H., G.W. and W.L. wrote the manuscript.

Corresponding authors

Correspondence to Guohao Wang, Wei Lu or Steven X. Hou.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks Geert Van Loo, Trent Woodruff and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 ARF1-deficient mice have movement defects.

Control (UBC-CreER/Arf1f/+, Arf1+/-) and ubiquitous ARF1-KO (UBC-CreER/Arf1f/f, Arf1-/-) mice were assayed. a, Schematic diagram of the experimental strategy used to generate ARF1-KO adult mice. b,c, Western blot analysis of ARF1 expression in different brain regions (b; Ctx-cerebral cortex, Str-striatum, BS-brain stem, MD-midbrain, OF-olfactory bulb) or different organs (c) from control (Arf1+/-) and the ubiquitous ARF1-KO (Arf1-/-) mice. d,e, Survival curve, body weight, forced swimming test and balance beam test of 2-month-old (d), 4-month-old (e) and 10-month-old (f) adult mice. n = 5 mice per group, one representative experiment from three independent experiments. g, Gait of control (Arf+/-) and ubiquitous ARF1-KO (Arf1-/-) mice was examined by footprinting assay at 2-month, 4-month, and 10-month of age. h, Quantification of stride lengths of front and hind footprints of mice in the indicated ages and control or ubiquitous ARF1-KO genotypes. n = 5 per group. Data are from one of three independent experiments and are represented as means ± SEM. P value was calculated using two-tailed t-test.

Source data

Extended Data Fig. 2 Muscle atrophy, demyelination and neuromuscular denervation of the ubiquitous ARF1-KO mice.

Control mice (UBC-CreER/Arf1f/f, Arf1+/-), the ubiquitous Arf1-KO (UBC-CreER/Arf1f/f, Arf1-/-). a, Masson’s trichrome stained sections of triangularis sterni (TS) muscles from 2-month-old the ubiquitous ARF1-KO comparison with control mice. Scale bar: 100 μm. b, Neurological score of 2-, 4- and 10-month-old control and the ubiquitous ARF1-KO mice. 2-m-old and 10-month-old: n = 5 mice per group, 3 male and 2 female mice. 4-month-old: n = 6 per group, 3 male and 3 female mice. c,d, Luxol Fast Blue Staining of cerebellum (c) and spinal cord (d) of control and the ubiquitous ARF1-KO mice. Scale bar: 100 μm. e, Immunofluorescence staining for MBP, CD3, CNP and Hoechst in spinal cords from control and the ubiquitous ARF1-KO mice. Scale bar: 10 μm. f, Immunofluorescence staining for PDGFRA, CC1 and Hoechst in cerebellum from control and the ubiquitous ARF1-KO mice. Scale bar: 100 μm (left), 20 μm (right). g, Immunoblot analysis of ARF1, myelin proteins and Neuron N (NeuN) in spinal cords of 2-, 4- and 10-month-old control and ubiquitous ARF1-KO mice. h, Syntaxin1 and α-bungarotoxin immunostaining of TS muscles from 2-month-old control and the ubiquitous ARF1-KO mice. Scale bar: 50 μm. Data are represented as mean with each replicate. Data represents one of three independent experiments. P value was calculated using two-tailed t-test.

Source data

Extended Data Fig. 3 Ablation of ARF1 in astrocytes, oligodendrocytes, myeloid cells and microglia did not show neurodegenerative phenotypes.

a-g, Body weight, balance beam test and neurological score were assayed in control and cell-type specifically ARF1-deleted mice. a-GFAP-CreER (astrocytes); Arf1f/+: n = 5 mice, 3 male and 2 female mice. Arf1f/f: n = 4 mice, 2 male and 2 female mice. b-Pdgfra-CreER (oligodendrocyte precursor); n = 6 mice, 3 male and 3 female mice. c-Plp1-CreER (oligodendrocytes and schwann cell); n = 6 mice, 3 male and 3 female mice. d-Sox10-CreER (oligodendrocyte lineage cell); n = 5 mice, 2 male and 3 female mice. e-LysM-CreER (myeloid cells); n = 4 mice, 2 male and 2 female mice in control group, and 2 male and 1 female mice in KO group. f-Cx3cr1-CreER (microglia); n = 4 mice, 2 male and 2 female mice in each group. g-Tmem119-CreER (microglia). n = 5 mice, 2 male and 3 female mice. One of three independent experiments was shown. Data are represented as mean ± SEM. h, Western blot for ARF1 expression was performed in the Cre-expressing cell types isolated from Arf1+/- and Arf1-/- mice. Data represents one of three independent experiments. Data are represented as mean with each replicate. P value was calculated using two-tailed t-test.

Source data

Extended Data Fig. 4 Neuronal specific ablation of ARF1 promotes formation of lipid droplets induces the neurodegenerative phenotypes in mice.

a-b, Masson’s trichrome (a), Syntaxin1 and α-bungarotoxin (b) immunostaining sections of triangularis sterni (TS) muscles from control and neuronal ARF1-KO mice. Scale bar: 50 μm. c, Luxol Fast Blue Staining of spinal cord from control and neuronal ARF1-KO mice. Scale bar: 50 μm. d, Immunoblot of ARF1, myelin proteins, GFAP, IBA1, and NeuN in spinal cords of control and neuronal ARF1-KO mice. e-f, Immunofluorescence (e) staining for PSD95, Synaptophysin (SYP) and Hoechst, and quantification of PSD95-positive and synaptophysin-positive synapses (f) in spinal cords from control and neuronal ARF1-KO mice. Scale bar: 50 μm (top), 20 μm (bottom). g-h, Representative spinal cord sections from mice with the control and neuronal ARF1-KO were analyzed by immunofluorescence staining (g) for NEFM & NEFH, IBA1, and Hoechst, and quantification of NEFM & NEFH dots (h). Scale bar: 50 μm (top), 10 μm (bottom). i, Electron microscopy (EM) images of axons in the medulla of control and neuronal ARF1-KO mice. Scale bar: 4 μm (top), 1 μm (top). j, Quantification of degenerated neurons in the EM sections. (e-j: n = 12 images observed from 3 mice). k, G-ratio analysis the EM images from the medulla of control and neuronal ARF1-KO mice. l, m, Immunofluorescence staining for IBA1, IL1β and Hoechst (l) and quantification of IL1β (m) in microglia from spinal cords of control and ubiquitous ARF1-KO mice. (n = 15 images of observation in 3 mice). n, o, Immunofluorescence staining for IBA1, ASC and Hoechst (n), and quantification of ASC in microglia (o) from spinal cords of control and ubiquitous ARF1-KO mice. (n = 20 images observed from 3 mice). l & n: Scale bar: 50 μm (left), 10 μm (middle and right). Data representative one of three independent experiments, data represented as mean ± SEM. P value was calculated by two-tailed t-test.

Source data

Extended Data Fig. 5 ARF1-ablated neurons promote formation of lipid droplets.

a, Electron microscopy showed lipid aggregates formulated in axons of neuronal ARF1-KO mice spinal cords. Scale bar: 500 nm. Data represents one of three independent experiments. b, c, Oil Red O staining (b) and quantification of Oil-Red-O-positive cells (c) of spinal cords from control and the neuronal ARF1-KO mice. Scale bar: 50 μm. Quantification of 20 images from three mice per group. Data are represented as mean ± SEM. d, Immunofluorescence stained with MitoSOXTM and MAP2 in primary culture neurons treated with Arf1 inhibitors GCA, BFA or DMSO. Scale bar: 10 μm. e-g, Treatment of control and the neuronal ARF1-KO mice with saline or mitochondrial ROS inhibitor MitoTempo (5 mg/kg) from day 6 to day 20 and checked the neurodegenerative phenotypes. n = 6 per group, 3 male and 3 female mice. Data representative one of three independent experiments. Data are represented as mean with each replicate. P value was calculated using two-tailed t-test in Fig.b, and Two-way ANOVA (or mixed model) with Tukey’s multiple comparisons test in Fig. e-g.

Source data

Extended Data Fig. 6 Lipid droplets exist in microglia but not astrocytes in the ARF1-deleted mice.

a,b, Immunofluorescence imaging (a) and quantification (b) of Bodipy-C11 in spinal cord of control and the ubiquitous Arf1-KO mice. (n = 15 images from 3 mice). Scale bar: 10 μm. c, d, Immunofluorescence imaging (c) and quantification (d) of BD-C11 and GFAP-positive astrocytes in spinal cord of control and neuronal ARF1-KO mice. (n = 20 images from 3 mice). Scale bar: 10 μm. e,f, Immunofluorescence staining (e) and quantification (f) of BD-C11 and IBA1-positive microglia in spinal cord of control and neuronal ARF1-KO mice. (n = 16 per group from 3 mice). Data are represented as mean ± SEM. g,h, Control (Arf1+/-) and neuronal ARF1-KO (Arf1-/-) mice were treated with PBS, AD4, and oxATP and assayed for various phenotypes: (g) neurological score and (h) balance beam test. (n = 5 mice per genotype). Data are represented as mean ± SEM with each replicate. i, Immunofluorescence staining for BD-C11 and Hoechst in spinal cord sections of control and neuronal Arf1-/- mice with indicated treatments. (n = 5 mice per condition), scale bars: 10 μm. j, MDA levels in spinal cord lysates from control and neuronal ARF1-KO mice with the indicated treatments (n = 5 per condition). k, quantification of peroxidated BD-C11 cells in neuronal Arf1-KO (Arf1-/-) mice and control mice with indicated treatments (n = 15 images observed from 3 mice). Data representative one of three independent experiments. Data are represented as mean ± SEM. P value in Fig. b, d, f, j, k using two-tailed t-test. P-value was calculated in Fig. g, h by Two-way ANOVA (or mixed model) with Tukey’s multiple comparisons test.

Source data

Extended Data Fig. 7 AD4 and oxATP significantly suppressed the phenotypes associated with ARF1 ablation.

Control (Thy1-CreER/Arf1f/+, Arf1+/-) and the neuronal ARF1-KO (Thy1-CreER/Arf1f/f, Arf1-/-) mice were treated with PBS, AD4, and oxATP and assayed for various phenotypes: a, Mitochondrial ROS was directly examined by using MitoSOX from mice with the indicated genotypes and treatments, then stained with MitoSOXTM (red) and MAP2 (green). Scale bars: 5 μm. Data represents one of three independent experiments. b,c, The synapses were checked by staining VGLUT1 and PSD95 (b) and quantified (c) in the spinal cord sections from mice with the indicated genotypes and treatments. (n = 20 images observed from 3 mice). Scale bars: 5 μm. d,e, Immunohistochemical staining (d) and quantification (e) for IBA1 and GFAP in spinal cords from mice with the indicated genotypes and treatments. Scale bar: 50 μm. (n = 8 images observed from 3 mice). f, Immunoblot of GSDMD, N-terminal GSDMD and Caspase11 in spinal cord lysates from control (Arf+/-) and neuronal ARF1-KO (Arf1-/-) mice. GAPDH was used as loading control. Data represents one of three independent experiments. g, The levels of several cytokines, complement C1q and C3 were measured by qRT-PCR in the mouse spinal cord lysates of control and neuronal ARF1-KO mice. (n = 4 mice in each group). h-j, The levels of Tnf and Il1α were measured by qRT-PCR from isolated microglia (h), Astrocytes (i), and neurons (j), in control and neuronal Arf1-KO mice. (n = 4 per group). k-n, The Levels of IL1α (k), IL1β (l), IL18 (m), TNF (n) were measured by ELISA in the mouse spinal cord lysates of control andneuronal ARF1-KO with treatments. (n = 5 per group). Data are represented one of three independent experiments and shown as mean ± SEM. P value was calculated by two-tailed t-test.

Source data

Extended Data Fig. 8 Knocking down Nlrp3 suppressed the neurodegenerative phenotypes of the neuronal ARF1-KO mice.

a-j, The defects of synapses in neuronal ARF1-KO mice were restored by MCC905 treatment (a, b) or NLRP3 knockout (c, d). Synapses were checked by staining of VGLUT1 and PSD95 (a, c) and quantified (b, d) in the spinal cord from mice with the indicated genotypes. Data represents one of three independent experiments. e-j, The MitoSOX signals were suppressed in the neuronal ARF1-KO mice after MCC950 treatment (e) or in NLRP3 knockout (f). However, the lipid peroxidation associated with neuronal ARF1-KO mice was not suppressed after MCC950 treatment (g, h) or NLRP3 knockout (i, j). Scale bars 5 μm. (b & d: n = 19, h & j: n = 6 images observed from 3 mice, one represented experiment from three independent replicates). k, Western blot of GPX4 in spinal cord of control (Arf1+/-) and neuronal ARF1-KO mice (Arf1-/-), β-ACTIN was used as a loading control. Data represents one of three independent experiments. l, experiment set up for ferroptosis inhibitor treatment of mice. m, Inhibition of ferroptosis did not alleviate the neurodegenerative phenotypes of neuronal ARF1-KO mice. (Arf1+/-+DMSO & Arf1-/- + Baicalein, n = 5 each group, 3 male 2 female mice; other groups: n = 6 per group, 3 male and 3 female mice). Data are represented one of three independent experiments and shown as mean ± SEM or mean with each replicate. P value was calculated using two-tailed t-test in Fig. b, d, h, j or two-way ANOVA (mixed model) with Tukey’s multiple comparisons test in Fig. m.

Source data

Extended Data Fig. 9 Inflammation relative genes transcriptional change in spinal cords of control and the ARF1-KO mice.

a–d, Genes transcriptional change in spinal cords of control and the neuronal ARF1-KO mice. Heatmaps that show differentially expressed genes with changes >2, >4 and p < 0.05 in spinal cords of control (Thy1-CreER/Arf1f/+, Arf1+/-) and neuronal ARF1-KO (Thy1-CreER/Arf1f/f, Arf1-/-) mice. (n = 3 per group). e, Heatmaps that show differentially expressed inflammasome-related genes with changes >1 in spinal cords of control (Thy1-CreER/Arf1f/+, Arf1+/-) and neuronal ARF1-KO (Thy1-CreER/Arf1f/f, Arf1-/-) mice. (n = 3 per group). f, Enriched ontology clusters in spinal cords of neuronal ARF1-KO mice. The most enriched clusters up-regulated genes are shown in neuronal ARF1-KO in comparison with control mice.

Extended Data Fig. 10 The most down-regulated and up-regulated gene groups in spinal cords of neuronal ARF1-KO mice.

a-d, The most down-regulated and up-regulated genes related to myelin and inflammatory responses are shown in neuronal ARF1-KO in comparison with control mice. Myelin sheath genes (a) and inflammatory response genes (c) were shown in spinal cord of neuronal ARF1-KO mice. The networks of Myelin sheath genes (b) and the inflammatory genes (d) through analyzing protein-protein interaction (PPI) using WebGestalt online software. e, Proposed model of ARF1-ablation-induced neurodegeneration.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Supplementary Video 1

Representative video of 2-month-old control mice (UBC-CreER/Arf1f/+; Arf1+/−) after tamoxifen injection for 22 d.

Supplementary Video 2

Representative video of 2-month-old ubiquitous ARF1-KO mice (UBC-CreER/Arf1f/f; Arf1−/−) after tamoxifen injection for 22 d.

Supplementary Video 3

Representative video of 2-month-old neuronal control mice (Thy1-CreER/Arf1f/+; Arf1+/−) after tamoxifen injection for 23 d.

Supplementary Video 4

Representative video of 2-month-old neuronal ARF1-KO mice (Thy1-CreER/Arf1f/f; Arf1−/−) after tamoxifen injection for 23 d.

Supplementary Video 5

Representative video of 2-month-old double neuronal ARF1 and NLRP3 knockout mice (Thy1-CreER/Arf1f/f/NLRP3/−; Arf1/−/NLRP3−/−) after tamoxifen injection for 22 d.

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, G., Yin, W., Shin, H. et al. Neuronal accumulation of peroxidated lipids promotes demyelination and neurodegeneration through the activation of the microglial NLRP3 inflammasome. Nat Aging 1, 1024–1037 (2021). https://doi.org/10.1038/s43587-021-00130-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00130-7

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