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:

Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models

Abstract

Defective brain hormonal signaling has been associated with Alzheimer’s disease (AD), a disorder characterized by synapse and memory failure. Irisin is an exercise-induced myokine released on cleavage of the membrane-bound precursor protein fibronectin type III domain-containing protein 5 (FNDC5), also expressed in the hippocampus. Here we show that FNDC5/irisin levels are reduced in AD hippocampi and cerebrospinal fluid, and in experimental AD models. Knockdown of brain FNDC5/irisin impairs long-term potentiation and novel object recognition memory in mice. Conversely, boosting brain levels of FNDC5/irisin rescues synaptic plasticity and memory in AD mouse models. Peripheral overexpression of FNDC5/irisin rescues memory impairment, whereas blockade of either peripheral or brain FNDC5/irisin attenuates the neuroprotective actions of physical exercise on synaptic plasticity and memory in AD mice. By showing that FNDC5/irisin is an important mediator of the beneficial effects of exercise in AD models, our findings place FNDC5/irisin as a novel agent capable of opposing synapse failure and memory impairment in AD.

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: CNS FNDC5/irisin is reduced in AD.
Fig. 2: Brain FNDC5/irisin is reduced in AD models.
Fig. 3: Downregulation of brain FNDC5/irisin impairs synaptic plasticity and object recognition memory in mice.
Fig. 4: Brain FNDC5/irisin rescues defective synaptic plasticity and memory in AD mice.
Fig. 5: Irisin triggers the brain cAMP–PKA–CREB signaling pathway.
Fig. 6: FNDC5/irisin mediates the beneficial effects of exercise on synaptic plasticity and memory.

Similar content being viewed by others

Data availability

Mass spectrometry raw data (Fig. 1 and Extended Data Fig. 1) and full blots (Fig. 2 and Extended Data Figs. 3,4, and 10) are available as source data files accompanying this article. Additional data that support the findings of this study are available from the corresponding author upon reasonable request. Requests of datasets obtained from human research will be subject to additional review steps by the IRB that has granted permit for a particular research. Please contact the corresponding authors for additional information.

References

  1. Prince, M. et al. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement. 9, 63–75.e62 (2013).

    Article  Google Scholar 

  2. Ferreira, S. T., Lourenco, M. V., Oliveira, M. M. & De Felice, F. G. Soluble amyloid-β oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front. Cell. Neurosci. 9, 191 (2015).

    PubMed  PubMed Central  Google Scholar 

  3. Fernandez, A. M. & Torres-Alemán, I. The many faces of insulin-like peptide signalling in the brain. Nat. Rev. Neurosci. 13, 225–239 (2012).

    Article  CAS  Google Scholar 

  4. Biessels, G. J. & Reagan, L. P. Hippocampal insulin resistance and cognitive dysfunction. Nat. Rev. Neurosci. 16, 660–671 (2015).

    Article  CAS  Google Scholar 

  5. McEwen, B. S. Preserving neuroplasticity: role of glucocorticoids and neurotrophins via phosphorylation. Proc. Natl Acad. Sci. USA 112, 15544–15545 (2015).

    CAS  PubMed  Google Scholar 

  6. During, M. J. et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat. Med. 9, 1173–1179 (2003).

    Article  CAS  Google Scholar 

  7. Chiu, S.-L., Chen, C.-M. & Cline, H. T. Insulin receptor signaling regulates synapse number, dendritic plasticity, and circuit function in vivo. Neuron 58, 708–719 (2008).

    Article  CAS  Google Scholar 

  8. Grillo, C. A. et al. Hippocampal insulin resistance impairs spatial learning and synaptic plasticity. Diabetes 64, 3927–3936 (2015).

    Article  CAS  Google Scholar 

  9. Irving, A. J. & Harvey, J. Leptin regulation of hippocampal synaptic function in health and disease. Philos. Trans. R. Soc. Lond. B 369, 20130155 (2013).

    Article  Google Scholar 

  10. Lourenco, M. V., Ferreira, S. T. & De Felice, F. G. Neuronal stress signaling and eIF2α phosphorylation as molecular links between Alzheimer’s disease and diabetes. Prog. Neurobiol. 129, 37–57 (2015).

    Article  CAS  Google Scholar 

  11. Bomfim, T. R. et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease-associated Aβ oligomers. J. Clin. Invest. 122, 1339–1353 (2012).

    Article  CAS  Google Scholar 

  12. Talbot, K. et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J. Clin. Invest. 122, 1316–1338 (2012).

    Article  CAS  Google Scholar 

  13. De Felice, F. G., Lourenco, M. V. & Ferreira, S. T. How does brain insulin resistance develop in Alzheimer’s disease? Alzheimers Dement. 10, S26–S32 (2014).

    Article  Google Scholar 

  14. Wadman, M. US government sets out Alzheimer’s plan. Nature 485, 426–427 (2012).

    Article  CAS  Google Scholar 

  15. De Felice, F. G. Alzheimer’s disease and insulin resistance: translating basic science into clinical applications. J. Clin. Invest. 123, 531–539 (2013).

    Article  CAS  Google Scholar 

  16. Boström, P. et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463–468 (2012).

    Article  Google Scholar 

  17. Jedrychowski, M. P. et al. Detection and quantitation of circulating human irisin by tandem mass spectrometry. Cell Metab. 22, 734–740 (2015).

    Article  CAS  Google Scholar 

  18. Wrann, C.D. et al. Exercise induces hippocampal BDNF through a PGC-1α/FNDC5 pathway. Cell Metab. 18, 649–659 (2013).

    Article  CAS  Google Scholar 

  19. Chen, K. et al. Irisin protects mitochondria function during pulmonary ischemia/reperfusion injury. Sci. Transl. Med. 9, eaao6298 (2017).

    Article  Google Scholar 

  20. Lee, P. et al. Irisin and FGF21 are cold-induced endocrine activators of brown fat function in humans. Cell Metab. 19, 302–309 (2014).

    Article  CAS  Google Scholar 

  21. Schumacher, M. A., Chinnam, N., Ohashi, T., Shah, R. S. & Erickson, H. P. The structure of irisin reveals a novel intersubunit β-sheet fibronectin type III (FNIII) dimer: implications for receptor activation. J. Biol. Chem. 288, 33738–33744 (2013).

    Article  CAS  Google Scholar 

  22. Mucke, L. & Selkoe, D. J. Neurotoxicity of amyloid β-protein: synaptic and network dysfunction. Cold Spring Harb. Perspect. Med. 2, a0063381 (2012).

    Article  Google Scholar 

  23. Sebollela, A. et al. Amyloid-β oligomers induce differential gene expression in adult human brain slices. J. Biol. Chem. 287, 7436–7445 (2012).

    Article  CAS  Google Scholar 

  24. Colaianni, G. et al. The myokine irisin increases cortical bone mass. Proc. Natl Acad. Sci. USA 112, 12157–12162 (2015).

    Article  CAS  Google Scholar 

  25. Figueiredo, C. P. et al. Memantine rescues transient cognitive impairment caused by high-molecular-weight Aβ oligomers but not the persistent impairment induced by low-molecular-weight oligomers. J. Neurosci. 33, 9626–9634 (2013).

    Article  CAS  Google Scholar 

  26. Lourenco, M. V. et al. TNF-α mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s β-amyloid oligomers in mice and monkeys. Cell Metab. 18, 831–843 (2013).

    Article  CAS  Google Scholar 

  27. Jankowsky, J. L. et al. Co-expression of multiple transgenes in mouse CNS: a comparison of strategies. Biomol. Eng. 17, 157–165 (2001).

    Article  CAS  Google Scholar 

  28. Cheng, A. et al. Involvement of PGC-1α in the formation and maintenance of neuronal dendritic spines. Nat. Commun. 3, 1250 (2012).

    Article  Google Scholar 

  29. Holcomb, L. et al. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat. Med. 4, 97–100 (1998).

    Article  CAS  Google Scholar 

  30. Ma, T. et al. Suppression of eIF2α kinases alleviates Alzheimer’s disease-related plasticity and memory deficits. Nat. Neurosci. 16, 1299–1305 (2013).

    Article  CAS  Google Scholar 

  31. Yang, W. et al. Repression of the eIF2α kinase PERK alleviates mGluR-LTD impairments in a mouse model of Alzheimer’s disease. Neurobiol. Aging 41, 19–24 (2016).

    Article  CAS  Google Scholar 

  32. Trinh, M. A. & Klann, E. Translational control by eIF2α kinases in long-lasting synaptic plasticity and long-term memory. Neurobiol. Learn. Mem. 105, 93–99 (2013).

    Article  CAS  Google Scholar 

  33. Gong, B. et al. Persistent improvement in synaptic and cognitive functions in an Alzheimer mouse model after rolipram treatment. J. Clin. Invest. 114, 1624–1634 (2004).

    Article  CAS  Google Scholar 

  34. Vitolo, O. V. et al. Amyloid β-peptide inhibition of the PKA/CREB pathway and long-term potentiation: reversibility by drugs that enhance cAMP signaling. Proc. Natl Acad. Sci. USA 99, 13217–13221 (2002).

    Article  CAS  Google Scholar 

  35. Schaefer, N. et al. The malleable brain: plasticity of neural circuits and behavior: a review from students to students. J. Neurochem. 142, 790–811 (2017).

    Article  CAS  Google Scholar 

  36. Katsnelson, A., De Strooper, B. & Zoghbi, H. Y. Neurodegeneration: from cellular concepts to clinical applications. Sci. Transl. Med. 8, 364ps318 (2016).

    Article  Google Scholar 

  37. Selkoe, D. J. Alzheimer’s disease is a synaptic failure. Science 298, 789–791 (2002).

    Article  CAS  Google Scholar 

  38. Lepeta, K. et al. Synaptopathies: synaptic dysfunction in neurological disorders: a review from students to students. J. Neurochem. 138, 785–805 (2016).

    Article  CAS  Google Scholar 

  39. Zhang, Y. et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes 63, 514–525 (2014).

    Article  CAS  Google Scholar 

  40. Timmons, J. A., Baar, K., Davidsen, P. K. & Atherton, P. J. Is irisin a human exercise gene? Nature 488, E9–E10 (2012).

    Article  CAS  Google Scholar 

  41. Albrecht, E. et al. Irisin: a myth rather than an exercise-inducible myokine. Sci. Rep. 5, 8889 (2015).

    Article  CAS  Google Scholar 

  42. Martin, K. C. & Kandel, E. R. Cell adhesion molecules, CREB, and the formation of new synaptic connections. Neuron 17, 567–570 (1996).

    Article  CAS  Google Scholar 

  43. Suwabe, K. et al. Rapid stimulation of human dentate gyrus function with acute mild exercise. Proc. Natl Acad. Sci. USA 115, 10487–10492 (2018).

    Article  CAS  Google Scholar 

  44. van Praag, H., Fleshner, M., Schwartz, M. W. & Mattson, M. P. Exercise, energy intake, glucose homeostasis, and the brain. J. Neurosci. 34, 15139–15149 (2014).

    Article  Google Scholar 

  45. Neufer, P. D. et al. Understanding the cellular and molecular mechanisms of physical activity-induced health benefits. Cell Metab. 22, 4–11 (2015).

    Article  CAS  Google Scholar 

  46. Baker, L. D. et al. Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch. Neurol. 67, 71–79 (2010).

    PubMed  PubMed Central  Google Scholar 

  47. Buchman, A. S. et al. Total daily physical activity and the risk of AD and cognitive decline in older adults. Neurology 78, 1323–1329 (2012).

    Article  CAS  Google Scholar 

  48. Okonkwo, O. C. et al. Physical activity attenuates age-related biomarker alterations in preclinical AD. Neurology 83, 1753–1760 (2014).

    Article  CAS  Google Scholar 

  49. Müller, S. Relationship between physical activity, cognition, and Alzheimer pathology in autosomal dominant Alzheimer’s disease. Alzheimers Dement. 14, 1427–1437 (2018).

    Article  Google Scholar 

  50. Mattson, M. P. Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab. 16, 706–722 (2012).

    Article  CAS  Google Scholar 

  51. Moon, H. Y. et al. Running-induced systemic cathepsin B secretion is associated with memory function. Cell Metab. 24, 332–340 (2016).

    Article  CAS  Google Scholar 

  52. Sleiman, S. F. et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. eLife 5, e15092 (2016).

    Article  Google Scholar 

  53. Smith, R. W., Wang, J., Bucking, C. P., Mothersill, C. E. & Seymour, C. B. Evidence for a protective response by the gill proteome of rainbow trout exposed to X-ray induced bystander signals. Proteomics 7, 4171–4180 (2007).

    Article  CAS  Google Scholar 

  54. Mendes, N. D. et al. Free-floating adult human brain-derived slice cultures as a model to study the neuronal impact of Alzheimer’s disease-associated Aβ oligomers. J. Neurosci. Meth. 307, 203–209 (2018).

    Article  CAS  Google Scholar 

  55. Drummond, C. et al. Deficits in narrative discourse elicited by visual stimuli are already present in patients with mild cognitive impairment. Front. Aging Neurosci. 7, 96 (2015).

    Article  Google Scholar 

  56. Ledo, J. H. et al. Amyloid-β oligomers link depressive-like behavior and cognitive deficits in mice. Mol. Psychiatry 18, 1053–1054 (2013).

    Article  CAS  Google Scholar 

  57. Ledo, J. H. et al. Cross talk between brain innate immunity and serotonin signaling underlies depressive-like behavior induced by Alzheimer’s amyloid-β oligomers in mice. J. Neurosci. 36, 12106–12116 (2016).

    Article  CAS  Google Scholar 

  58. Trinchese, F. et al. Inhibition of calpains improves memory and synaptic transmission in a mouse model of Alzheimer disease. J. Clin. Invest. 118, 2796–2807 (2008).

    Article  CAS  Google Scholar 

  59. Alamed, J., Wilcock, D. M., Diamond, D. M., Gordon, M. N. & Morgan, D. Two-day radial-arm water maze learning and memory task; robust resolution of amyloid-related memory deficits in transgenic mice. Nat. Protoc. 1, 1671–1679 (2006).

    Article  CAS  Google Scholar 

  60. Puzzo, D. et al. Phosphodiesterase 5 inhibition improves synaptic function, memory, and amyloid-β load in an Alzheimer’s disease mouse model. J. Neurosci. 29, 8075–8086 (2009).

    Article  CAS  Google Scholar 

  61. Madeira, C. et al. d-serine levels in Alzheimer’s disease: implications for novel biomarker development. Transl. Psychiatry 5, e561 (2015).

    Article  CAS  Google Scholar 

  62. Seixas Da Silva, G. S. et al. Amyloid-β oligomers transiently inhibits AMP-activated kinase and cause metabolic defects in hippocampal neurons. J. Biol. Chem. 292, 7395–7406 (2017).

    Article  CAS  Google Scholar 

  63. Gong, B. et al. Ubiquitin hydrolase Uch-L1 rescues β-amyloid-induced decreases in synaptic function and contextual memory. Cell 126, 775–788 (2006).

    Article  CAS  Google Scholar 

  64. De Felice, F. G. et al. Aβ oligomers induce neuronal oxidative stress through an N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. J. Biol. Chem. 282, 11590–11601 (2007).

    Article  CAS  Google Scholar 

  65. Lambert, M. P. et al. Monoclonal antibodies that target pathological assemblies of Aβ. J. Neurochem. 100, 23–35 (2007).

    Article  CAS  Google Scholar 

  66. Abràmoff, M. D., Magalhães, P. J. & Ram, S. J. Image processing with ImageJ. Biophotonics Int. 11, 36–42 (2004).

    Google Scholar 

  67. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔC T method. Methods 25, 402–408 (2001).

    Article  CAS  Google Scholar 

  68. Brito-Moreira, J. et al. Interaction of amyloid-β (Aβ) oligomers with neurexin 2α and neuroligin 1 mediates synapse damage and memory loss in mice. J. Biol. Chem. 292, 7327–7337 (2017).

    Article  CAS  Google Scholar 

  69. Schmidt, E. K., Clavarino, G., Ceppi, M. & Pierre, P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat. Methods 6, 275–277 (2009).

    Article  CAS  Google Scholar 

  70. Beckman, D. et al. Prion protein modulates monoaminergic systems and depressive-like behavior in mice. J. Biol. Chem. 290, 20488–20498 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from Alzheimer Society of Canada (to F.G.D.F.) and the Weston Brain Institute (to F.G.D.F.), National Institute for Translational Neuroscience (INNT/Brazil) (465346/2014-6 to S.T.F. and F.G.F.), Human Frontier Science Program (to F.G.D.F.), International Society for Neurochemistry (CAEN 1B to M.V.L.), National Institutes of Health (NIH-R01NS049442 to O.A.), Canadian Institutes of Health Research (CIHR MOP 136940 and MOP 89919 to V.F.P. and M.A.M.P.), and from the Brazilian funding agencies Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) (451195/2017-5 to M.V.L., 406436/2016-9 to S.T.F., and 473324/2013-0 to F.G.D.F.) and Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) (202.817/2016 to M.V.L., 201.432/2014 to S.T.F., and 202.944/2015 to F.G.D.F.). R.L.F, G.B.d.F., G.C.K., F.C.R., J.R.C., D.B., and L.F.-G. were supported by fellowships granted by FAPERJ, CNPq, or Comissão de Aperfeiçoamento de Pessoal de Nível Superior (CAPES/Brazil; financial code 001). S.M. was supported by an NIH T32 grant (AG057461). We acknowledge the University of Kentucky Alzheimer’s Disease Center and its Neuropathology Core, which is supported by NIH/NIA P30 AG028383, for brain samples. We thank W.L. Klein (Northwestern University) for the kind gift of oligomer-specific NU4 antibodies, B.M. Spiegelman (Harvard University) for sharing AdGFP and AdFNDC5 adenoviral constructs, and J. Wang (Queen’s University, Canada) for help with mass spectrometry analyses. We also thank A. Lepelley, M. Oliveira, M. Melo, A.C. Rangel, and the CENABIO team for technical and/or administrative assistance.

Author information

Authors and Affiliations

Authors

Contributions

M.V.L., R.L.F., O.A., S.T.F., and F.G.D.F. designed the study. M.V.L., R.L.F., G.B.d.F., H.Z., G.C.K, F.C.R., R.A.G., J.R.C., D.B., A.S., H.B., L.A.G., L.F.-G., S.M., and J.F.A. performed the research. M.V.L., R.L.F., G.B.d.F., H.Z., D.B., A.S., S.M., J.F.A., O.A., S.T.F., and F.G.D.F. analyzed the data. J.F.A., D.M.W., J.M.d.S., S.A.-L., V.F.P., M.A.M.P., F.T.-M., P.M., and O.A. contributed the reagents, materials, and analysis tools. M.V.L., R.L.F., G.B.d.F., A.S, J.F.A., F.T-M., P.M., O.A., S.T.F., and F.G.D.F. analyzed and discussed the results. M.V.L., R.L.F., S.T.F., and F.G.D.F. wrote the manuscript.

Corresponding authors

Correspondence to Ottavio Arancio, Sergio T. Ferreira or Fernanda G. De Felice.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Validation of anti-FNDC5 for detection of brain FNDC5/irisin.

a, Antibody blocking assay. Previous incubation of anti-FNDC5 (Abcam; ab131390) with increasing molar ratios of recombinant irisin (Adipogen; AG-40B-0136) reduces the signal of recombinant irisin detected by immunoblotting. The experiment was repeated 4 times with similar results. b, Left, Immunoblot of mouse hippocampus homogenate probed with anti-FNDC5. Immunolabeled bands 1–4 (see Results) were used to guide excision of the corresponding bands from the other half of the SDS–PAGE gel that had not been electroblotted (right). Band excision was guided by overlaying the unstained gel onto an image of the developed immunoblot (right panel). Excised bands were subjected to in-gel tryptic digestion followed by mass spectrometry analysis, as described in the Methods. Peptides identified by mass spectrometry in each band are indicated. c, Full-length FNDC5 amino acid sequence. The sequence corresponding to irisin is underlined. Peptides identified by mass spectrometry in excised bands are highlighted in green (bands 1, 2 and 3) or blue (band 4). d, Antibody used in the Phoenix ELISA kit (EK-067-29) recognizes recombinant irisin expressed in CHO cells (Adipogen; AG-40B-0136). The experiment was repeated 3 times with similar results. See Source Data 1 for original data.

Extended Data Fig. 2 CSF and plasma irisin correlations.

a,b, CSF:plasma irisin levels correlation in controls (N = 26) (a) or AD patients (b) (N = 14; lines represent linear regression fits to the data; r2 and P values as indicated in the figure). c, CSF to plasma irisin ratio is selectively reduced in AD patients, as compared to controls, MCI or LBD patients (N = 26 controls, 14 MCI, 11 AD, 13 LBD cases). Data are shown as mean ± s.e.m., *P < 0.05; One-way ANOVA with Holm-Sidak post-test.

Extended Data Fig. 3 AβOs reduce FNDC5/irisin levels in hippocampal neurons.

ac, Primary cultured hippocampal neurons were exposed to 500 nM AβOs for 24 h. Fndc5 mRNA (a) and FNDC5/irisin protein levels (b,c) in cultured hippocampal neurons exposed or not to AβOs (N = 4 experiments with independent neuronal cultures and AβO preparations; data are shown as mean ± s.e.m., **P < 0.01; paired Student’s t-test; two-sided). See Source Data 5 for original data. d,e, Colocalization of surface FNDC5 immunoreactivity (green) with β–tubulin III (red) immunoreactivity in cultured hippocampal neurons (d). Colocalization of surface FNDC5 immunoreactivity (green) with glial fibrillary acidic protein (GFAP) (red) (e). The experiment was repeated 2 times with similar results in independent cultures. f,g, Primary cultured hippocampal neurons were exposed to 500 nM AβOs for 24 h. g, Summary quantification of surface FNDC5 immunoreactivity in cultured neurons (N = 4 experiments with independent neuronal cultures and AβO preparations; 30 images (from 2–3 coverlips) per condition per experiment. Data are shown as mean ± s.e.m. * P < 0.05, paired Student’s t-test; two-sided). h, Surface FNDC5 immunoreactivity (red) in 18 DIV cultured hippocampal neurons after lentiviral knockdown of FNDC5 (shFNDC5) (N = 2 experiments with independent cultures; 30 images (from 2–3 coverslips) per condition per experiment). Scale bar = 10 mm.

Source data

Extended Data Fig. 4 AβOs reduce hippocampal FNDC5, PGC-1α, and PPARγ expression.

a, Hippocampal, cortical, and skeletal muscle (gastrocnemius) expression of FNDC5 in C57BL/6 mice (N = 6 per group). Data are shown as mean ± s.e.m.; *P < 0.05; paired one-way ANOVA with Holm-Sidak correction; two-sided. b,c, Hippocampal FNDC5 mRNA in C57BL/6 i.c.v-infused with 10 pmol AβOs for 24 h (N = 8 per group) (b) or 7 days (N = 5 per group) (c). Data are shown as mean ± s.e.m.; *P < 0.05; Student’s t-test; two-sided). d,e, Skeletal muscle (gastrocnemius) FNDC5 mRNA in C57BL/6 i.c.v-infused with 10 pmol AβOs for 24 h (N = 7 Veh, 5 AβOs) (d) or 7 days (N = 7 Veh, 4 AβOs) (e). Data are shown as mean ± s.e.m.; *P < 0.05; Student’s t-test; two-sided). fk, Hippocampal mRNA levels of PGC-1α (f,h), PPARγ (g,i) and PPARα (j,k) were measured 24 h or 7 days after infusion, as indicated (N = 5 per group). mRNA levels were normalized by β-actin expression. (l,m) AβOs reduced hippocampal PGC-1α protein levels in C57BL/6 mice 7 days after infusion (N = 7 Veh, 6 AβOs). Data are shown as mean ± s.e.m.; *P < 0.05; Student’s t-test; two-sided). See Source Data 6 for original data.

Source data

Extended Data Fig. 5 Lentiviral vectors expressing shFNDC5 did not cause motor impairment or affect body weight gain in mice.

a,b, Distance traveled (a) and mean velocity (b) of mice allowed to explore an open field arena for 5 min (N = 9 mice for shCtrl, 10 mice each for shFNDC5 (1) and (2) groups). Data are shown as mean ± s.e.m. c, Body weight measured 0, 14 or 28 days after lentiviral injections (N = 10 mice per group; one-way ANOVA followed by Holm-Sidak post-test). Data are shown as mean ± s.e.m. No significantly statistical difference was found among groups.

Extended Data Fig. 6 Intrahippocampal administration of recombinant irisin prevents AβO-induced memory impairment.

a,b, Male Swiss mice (3 months old) were bilaterally injected with recombinant irisin (75 pmol per hippocampus) and received 10 pmol AβOs i.c.v. Novel object recognition (a) and contextual fear conditioning (b) tasks (tested 5 days post-infusion of AβOs) (N = 9 mice for Veh, 10 for AβOs, 9 for irisin and 7 for AβOs + irisin). Data are shown as mean ± s.e.m. *P < 0.05, two-way ANOVA with Holm-Sidak post-test; two-sided.

Extended Data Fig. 7 Validation of PCR array results by qPCR.

af, AdFNDC5 expression of 6 synapse plasticity-related genes after AβO injection: Egr1 (a), Egr4 (b), Gria2 (c), Grm2 (d), Nptx2 (e), and Ppp3ca (f). N = 6 mice per experimental group. Data are shown as mean ± s.e.m. *P < 0.05; two-way ANOVA with Holm-Sidak post-test; two-sided.

Extended Data Fig. 8 FNDC5/irisin rescues defective synaptic plasticity and memory in APP/PS1 M146L mice.

ad, APP/PS1 M146L mice (or WT littermates) were injected i.c.v. with an adenoviral vector expressing full-length FNDC5 (AdFNDC5) or green fluorescent protein (AdGFP, used as a control). Hippocampal slices were obtained and subjected to high-frequency stimulation for LTP recordings. Field excitatory postsynaptic potentials (fEPSP) in hippocampal slices from each experimental group (N = 9 slices for vehicle AdGFP, 7 for APP/PS1 M146L AdGFP, 8 for WT AdFNDC5, 7 for APP/PS1 M146L AdFNDC5; from 3–4 animals per group) (a). fEPSP at 120 min (b). I.c.v.-injected AdFNDC5 rescued memory impairment in 3–4 months-old APP/PS1 M146L mice in 2-day radial arm water maze (c) and in contextual fear conditioning (d) (N = 10 mice for WT GFP, 8 for APP/PS1 GFP, 11 for WT FNDC5, 10 for APP/PS1 FNDC5). Data are shown as mean ± s.e.m. *P < 0.05; two-way ANOVA with Holm-Sidak post-test; two-sided.

Extended Data Fig. 9 Irisin counteracts AD-linked activation of cellular stress response and dendritic spine loss in hippocampal neurons.

a, Effect of irisin on AβO-induced increases in eIF2α-P (green) and upregulation of nuclear ATF4 (red). Nuclei were counterstained in blue (DAPI). Scale bar = 5 mm. b,c, Summary quantification of immunocytochemistry experiments (N = 4 experiments with independent neuronal cultures and AβO preparations). *P < 0.05; two-way ANOVA with Holm-Sidak correction; two-sided. Data are represented by mean ± s.e.m. d,e, Summary quantification of of protein synthesis in hippocampal neurons, as measured by non-radioactive puromycin incorporation (SUnSET) normalized by β-actin levels (N = 4 experiments with independent hippocampal cultures and AβO preparations). *P < 0.05; two-way ANOVA with Holm-Sidak correction; two-sided. Data are represented by mean ± s.e.m. f,g, Representative images of dendritic spines in hippocampal neurons, as measured by F-actin labeling with Alexa-conjugated phalloidin (N = 5 experiments with independent neuronal cultures and AβO preparations). Scale bar = 20 mm. *P < 0.05, two-way ANOVA. Data are shown as mean ± s.e.m. At least 30 neurons were analyzed per condition per experiment in immunocytochemistry experiments. h, AβO binding to cultured hippocampal neurons, as detected by AβO-sensitive antibody NU4 (red), after treatment with recombinant irisin (25 nM). Scale bar, = 10 mm. The experiments were repeated 5 times with similar results. i, Summary quantification of 5 experiments with independent neuronal cultures and AβO preparations. Data are shown as mean ± s.e.m. *P < 0.05; paired one-way ANOVA; two-sided. j, AβO interaction with different proteins in a plate-binding assay. BSA was used as a negative control, while neuroligin-1 was used as a positive control (N = 3 experiments with independent AβO preparations)68. Representative dots were cropped from the same film. See Source Data 7 for original data. k, Double immunocytochemistry colocalization between AβOs (red) and surface FNDC5 (green) in primary cultured hippocampal neurons (3 experiments with independent neuronal cultures and AβO preparations, with 20–25 images (from 2–3 coverslips) per experiment). Scale bar = 5 mm. The experiments were repeated 3 times with similar results. lo, Levels of soluble (l,n) and insoluble Aβ42 (m,o) in hippocampus (N = 3 for AdGFP, 5 for AdFNDC5) and cortex (N = 3 for AdGFP, 4 for AdFNDC5) of APP/PS1 M146L mice. Data are shown as mean ± s.e.m. *P < 0.05; Student’s t-test; two-sided.

Source data

Extended Data Fig. 10 Exercise blocks AβO-induced memory impairment in mice.

Effect of exercise (swimming; 5 weeks, 5 days/week, 1 h/day) on memory impairment induced by i.c.v. infusion of AβOs in Swiss mice. ac, Novel object recognition assessed 24 h (N = 9 mice for vehicle, 8 for AβOs, 8 for exercise, 9 for exercise + AβOs) (a) or 5 days post-infusion of AβOs (N = 8 mice for vehicle, 10 for AβOs, 8 for exercise and 8 for exercise + AβOs) (b). Data are shown as mean ± s.e.m. *P < 0.05; one-sample Student’s t-test. Contextual fear conditioning (c) assessed 7 days post-infusion of AβOs (N = 7 mice for vehicle, 8 for AβOs, 9 for exercised and 6 for exercised + AβOs). *P > 0.05; two-way ANOVA followed by Holm-Sidak post-test; two-sided). d,e, Hippocampal FNDC5 mRNA (d) (N = 9 mice per group) and FNDC5/irisin protein levels (e; measured by ELISA) after 5 weeks of exercise (N = 6 mice each for vehicle, AβOs and exercise + AβOs, and 5 for exercise) in male Swiss mice that received 10 pmol AβOs i.c.v. f,g, Hippocampal BDNF levels in exercised mice (N = 10 mice for vehicle and AβOs, 11 for exercise, 13 for exercise + AβOs). Data are shown as mean ± s.e.m. *P < 0.05; **P < 0.01, two-way ANOVA followed by Holm-Sidak post-test; two-sided. Representative bands were cropped from the same membrane. See Source Data 8 for original data.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, describing patient demographics and PCR primer sequences.

Reporting Summary

Source data

Source data, Fig. 1

Mass spectrometry data for the corresponding figure.

Source data, Fig. 1

Western blots for the corresponding figures

Source data, Fig. 2

Western blots for the corresponding figures

Source data, Fig. 2

Western blots for the corresponding figures

Source data, Extended Data Fig. 3

Western blots for the corresponding figures

Source data, Extended Data Fig. 4

Western blots for the corresponding figures

Source data, Extended Data Fig. 9

Western blots for the corresponding figures

Source data, Extended Data Fig. 10

Western blots for the corresponding figures

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lourenco, M.V., Frozza, R.L., de Freitas, G.B. et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nat Med 25, 165–175 (2019). https://doi.org/10.1038/s41591-018-0275-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0275-4

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