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 hormone irisin is a critical regulator of cognitive function

An Author Correction to this article was published on 07 October 2021

This article has been updated

Abstract

Identifying secreted mediators that drive the cognitive benefits of exercise holds great promise for the treatment of cognitive decline in ageing or Alzheimer’s disease (AD). Here, we show that irisin, the cleaved and circulating form of the exercise-induced membrane protein FNDC5, is sufficient to confer the benefits of exercise on cognitive function. Genetic deletion of Fndc5/irisin (global Fndc5 knock-out (KO) mice; F5KO) impairs cognitive function in exercise, ageing and AD. Diminished pattern separation in F5KO mice can be rescued by delivering irisin directly into the dentate gyrus, suggesting that irisin is the active moiety. In F5KO mice, adult-born neurons in the dentate gyrus are morphologically, transcriptionally and functionally abnormal. Importantly, elevation of circulating irisin levels by peripheral delivery of irisin via adeno-associated viral overexpression in the liver results in enrichment of central irisin and is sufficient to improve both the cognitive deficit and neuropathology in AD mouse models. Irisin is a crucial regulator of the cognitive benefits of exercise and is a potential therapeutic agent for treating cognitive disorders including AD.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Genetic deletion of irisin impairs cognitive function in exercise and ageing.
Fig. 2: Pattern separation is impaired in F5KO mice and can be rescued by delivering irisin directly into the DG.
Fig. 3: Adult-born neurons, but not mature granule cells, in the hippocampus display aberrant activation in global F5KO mice.
Fig. 4: Adult-born neurons in the hippocampus develop abnormally in global F5KO mice.
Fig. 5: The transcriptome of adult-born neurons in the hippocampus is altered in global F5KO mice.
Fig. 6: Peripheral irisin improves cognitive function in transgenic mouse models of AD.
Fig. 7: Peripheral irisin reduces glia activation in transgenic mouse models of AD.
Fig. 8: Peripherally delivered irisin crosses the blood–brain barrier.

Similar content being viewed by others

Data availability

RNA-seq datasets are available at the Gene Expression Omnibus repository under accession number GSE174212, which contains the GSE174210, GSE174211 and GSE179078 datasets. Markers for astrocytes and microglia are available from http://dropviz.org. Data from the MSSM study are available from https://www.synapse.org/. The single-cell data are available from http://linnarssonlab.org/dentate/. Source data are provided with this paper.

Code availability

The code for the RNA-seq data analysis from the MSSM study is available in the Supplementary Information.

Change history

References

  1. Maass, A. et al. Vascular hippocampal plasticity after aerobic exercise in older adults. Mol. Psychiatry 20, 585–593 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Colcombe, S. & Kramer, A. F. Fitness effects on the cognitive function of older adults: a meta-analytic study. Psychol. Sci. 14, 125–130 (2003).

    Article  PubMed  Google Scholar 

  3. Voss, M. W. et al. Exercise and hippocampal memory systems. Trends Cogn. Sci. 23, 318–333 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. 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  PubMed  PubMed Central  Google Scholar 

  5. van Praag, H., Christie, B. R., Sejnowski, T. J. & Gage, F. H. Running enhances neurogenesis, learning and long-term potentiation in mice. Proc. Natl Acad. Sci. USA 96, 13427–13431 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Choi, S. H. et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science 361, (2018).

  7. Nichol, K. E. et al. Exercise alters the immune profile in Tg2576 Alzheimer mice toward a response coincident with improved cognitive performance and decreased amyloid. J. Neuroinflammation 5, 13 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kim, H. et al. Irisin mediates effects on bone and fat via αV integrin receptors. Cell 175, 1756–1768 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lourenco, M. V. et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nat. Med. 25, 165–175 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mustroph, M. L. et al. Aerobic exercise is the critical variable in an enriched environment that increases hippocampal neurogenesis and water maze learning in male C57BL/6J mice. Neuroscience 219, 62–71 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Kobilo, T. et al. Running is the neurogenic and neurotrophic stimulus in environmental enrichment. Learn. Mem. 18, 605–609 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bolz, L., Heigele, S. & Bischofberger, J. Running improves pattern separation during novel object recognition. Brain Plast. 1, 129–141 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  16. van Praag, H., Shubert, T., Zhao, C. & Gage, F. H. Exercise enhances learning and hippocampal neurogenesis in aged mice. J. Neurosci. 25, 8680–8685 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  17. McNaughton, B. L., Barnes, C. A., Meltzer, J. & Sutherland, R. J. Hippocampal granule cells are necessary for normal spatial learning but not for spatially selective pyramidal cell discharge. Exp. Brain Res. 76, 485–496 (1989).

    Article  CAS  PubMed  Google Scholar 

  18. Garthe, A. & Kempermann, G. An old test for new neurons: refining the Morris water maze to study the functional relevance of adult hippocampal neurogenesis. Front. Neurosci. 7, 63 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  19. McHugh, T. J. et al. Dentate gyrus NMDA receptors mediate rapid pattern separation in the hippocampal network. Science 317, 94–99 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Hunsaker, M. R. & Kesner, R. P. Evaluating the differential roles of the dorsal dentate gyrus, dorsal CA3 and dorsal CA1 during a temporal ordering for spatial locations task. Hippocampus 18, 955–964 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Clelland, C. D. et al. A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science 325, 210–213 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Creer, D. J., Romberg, C., Saksida, L. M., van Praag, H. & Bussey, T. J. Running enhances spatial pattern separation in mice. Proc. Natl Acad. Sci. USA 107, 2367–2372 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ally, B. A., Hussey, E. P., Ko, P. C. & Molitor, R. J. Pattern separation and pattern completion in Alzheimer’s disease: evidence of rapid forgetting in amnestic mild cognitive impairment. Hippocampus 23, 1246–1258 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Déry, N. et al. Adult hippocampal neurogenesis reduces memory interference in humans: opposing effects of aerobic exercise and depression. Front. Neurosci. 7, 66 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Sahay, A. et al. Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature 472, 466–470 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Besnard, A., Miller, S. M. & Sahay, A. Distinct dorsal and ventral hippocampal CA3 outputs govern contextual fear discrimination. Cell Rep. 30, 2360–2373 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rhodes, J. S., Garland, T. Jr. & Gammie, S. C. Patterns of brain activity associated with variation in voluntary wheel-running behavior. Behav. Neurosci. 117, 1243–1256 (2003).

    Article  PubMed  Google Scholar 

  28. Oladehin, A. & Waters, R. S. Location and distribution of Fos protein expression in rat hippocampus following acute moderate aerobic exercise. Exp. Brain Res. 137, 26–35 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Sahay, A., Wilson, D. A. & Hen, R. Pattern separation: a common function for new neurons in hippocampus and olfactory bulb. Neuron 70, 582–588 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Aimone, J. B., Deng, W. & Gage, F. H. Adult neurogenesis: integrating theories and separating functions. Trends Cogn. Sci. 14, 325–337 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Colgin, L. L., Moser, E. I. & Moser, M. B. Understanding memory through hippocampal remapping. Trends Neurosci. 31, 469–477 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Tapia-Rojas, C., Aranguiz, F., Varela-Nallar, L. & Inestrosa, N. C. Voluntary running attenuates memory loss, decreases neuropathological changes and induces neurogenesis in a mouse model of Alzheimer’s disease. Brain Pathol. 26, 62–74 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Moreno-Jiménez, E. P. et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 25, 554–560 (2019).

    Article  PubMed  Google Scholar 

  34. Spalding, K. L. et al. Dynamics of hippocampal neurogenesis in adult humans. Cell 153, 1219–1227 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tobin, M. K. et al. Human hippocampal neurogenesis persists in aged adults and Alzheimer’s disease patients. Cell Stem Cell 24, 974–982 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mu, Y. & Gage, F. H. Adult hippocampal neurogenesis and its role in Alzheimer’s disease. Mol. Neurodegener. 6, 85 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  37. McAvoy, K. M. et al. Modulating neuronal competition dynamics in the dentate gyrus to rejuvenate aging memory circuits. Neuron 91, 1356–1373 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhao, C., Jou, J., Wolff, L. J., Sun, H. & Gage, F. H. Spine morphogenesis in newborn granule cells is differentially regulated in the outer and middle molecular layers. J. Comp. Neurol. 522, 2756–2766 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Zhao, C., Teng, E. M., Summers, R. G. Jr., Ming, G. L. & Gage, F. H. Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus. J. Neurosci. 26, 3–11 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Piatti, V. C. et al. The timing for neuronal maturation in the adult hippocampus is modulated by local network activity. J. Neurosci. 31, 7715–7728 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. van Praag, H. et al. Functional neurogenesis in the adult hippocampus. Nature 415, 1030–1034 (2002).

    Article  PubMed  Google Scholar 

  42. Goncalves, J. T. et al. In vivo imaging of dendritic pruning in dentate granule cells. Nat. Neurosci. 19, 788–791 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Anacker, C. et al. Hippocampal neurogenesis confers stress resilience by inhibiting the ventral dentate gyrus. Nature 559, 98–102 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Duman, J. G. et al. The adhesion-GPCR BAI1 shapes dendritic arbors via Bcr-mediated RhoA activation causing late growth arrest. eLife 8, e47566 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Saunders, A. et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell 174, 1015–1030 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Vivar, C. et al. Monosynaptic inputs to new neurons in the dentate gyrus. Nat. Commun. 3, 1107 (2012).

    Article  PubMed  Google Scholar 

  47. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  PubMed  Google Scholar 

  48. Huang da, W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  PubMed  Google Scholar 

  49. Subramanian, A. et al. Gene-set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Jankowsky, J. L. et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum. Mol. Genet. 13, 159–170 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Fang, E. F. et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 22, 401–412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wan, Y.-W. et al. Meta-analysis of the Alzheimer’s disease human brain transcriptome and functional dissection in mouse models. Cell Rep. 32, 107908 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Haass, C. & Selkoe, D. J. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid β-peptide. Nat. Rev. Mol. Cell Biol. 8, 101–112 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Stakos, D. A. et al. The Alzheimer’s disease amyloid-beta hypothesis in cardiovascular aging and disease: JACC Focus Seminar. J. Am. Coll. Cardiol. 75, 952–967 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Oakley, H. et al. Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 26, 10129–10140 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pluvinage, J. V. & Wyss-Coray, T. Systemic factors as mediators of brain homeostasis, ageing and neurodegeneration. Nat. Rev. Neurosci. 21, 93–102 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Leyns, C. E. G. & Holtzman, D. M. Glial contributions to neurodegeneration in tauopathies. Mol. neurodegener. 12, 50 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Hochgerner, H., Zeisel, A., Lonnerberg, P. & Linnarsson, S. Conserved properties of dentate gyrus neurogenesis across postnatal development revealed by single-cell RNA sequencing. Nat. Neurosci. 21, 290–299 (2018).

    Article  CAS  PubMed  Google Scholar 

  60. 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 

  61. Guo, W. et al. Ablation of Fmrp in adult neural stem cells disrupts hippocampus-dependent learning. Nat. Med. 17, 559–565 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Guo, W., Patzlaff, N. E., Jobe, E. M. & Zhao, X. Isolation of multipotent neural stem or progenitor cells from both the dentate gyrus and subventricular zone of a single adult mouse. Nat. Protoc. 7, 2005–2012 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Sah, N., Peterson, B. D., Lubejko, S. T., Vivar, C. & van Praag, H. Running reorganizes the circuitry of one-week-old adult-born hippocampal neurons. Sci. Rep. 7, 10903 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Nuber, S. et al. Abrogating native α-synuclein tetramers in mice causes a L-DOPA-responsive motor syndrome closely resembling Parkinson’s disease. Neuron 100, 75–90 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Liu, B. et al. Space-like 56Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wild-type and Alzheimer’s-like transgenic mice. Sci. Rep. 9, 12118 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Caldarone, B. J. et al. The novel triple reuptake inhibitor JZAD-IV-22 exhibits an antidepressant pharmacological profile without locomotor stimulant or sensitization properties. J. Pharmacol. Exp. Ther. 335, 762–770 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Shi, Q. et al. Complement C3-deficient mice fail to display age-related hippocampal decline. J. Neurosci. 35, 13029–13042 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Morris, R. Developments of a water-maze procedure for studying spatial learning in the rat. J. Neurosci. Methods 11, 47–60 (1984).

    Article  CAS  PubMed  Google Scholar 

  69. Vorhees, C. V. & Williams, M. T. Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat. Protoc. 1, 848–858 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Leger, M. et al. Object recognition test in mice. Nat. Protoc. 8, 2531–2537 (2013).

    Article  CAS  PubMed  Google Scholar 

  71. Wolf, A., Bauer, B., Abner, E. L., Ashkenazy-Frolinger, T. & Hartz, A. M. A comprehensive behavioral test battery to assess learning and memory in 129S6/Tg2576 mice. PLoS ONE 11, e0147733 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Faizi, M. et al. Thy1-hAPP(Lond/Swe+) mouse model of Alzheimer’s disease displays broad behavioral deficits in sensorimotor, cognitive and social function. Brain Behav. 2, 142–154 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Zhao, X. & van Praag, H. Steps towards standardized quantification of adult neurogenesis. Nat. Commun. 11, 4275 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Young, K. & Morrison, H. Quantifying microglia morphology from photomicrographs of immunohistochemistry prepared tissue using ImageJ. J. Vis. Exp. 136, (2018).

  75. Fontaine, C. J. et al. Impaired bidirectional synaptic plasticity in juvenile offspring following prenatal ethanol exposure. Alcohol. Clin. Exp. Res. 43, 2153–2166 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Peñasco, S. et al. Endocannabinoid long-term depression revealed at medial perforant path excitatory synapses in the dentate gyrus. Neuropharmacology 153, 32–40 (2019).

    Article  PubMed  Google Scholar 

  77. Schmittgen, T. D. & Livak, K. J. Analyzing real-time PCR data by the comparative CT method. Nat. Protoc. 3, 1101–1108 (2008).

    Article  CAS  PubMed  Google Scholar 

  78. Schmider, A. B. et al. Two- and three-color STORM analysis reveals higher-order assembly of leukotriene synthetic complexes on the nuclear envelope of murine neutrophils. J. Biol. Chem. 295, 5761–5770 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Bilsland, J. G. et al. Behavioral and neurochemical alterations in mice deficient in anaplastic lymphoma kinase suggest therapeutic potential for psychiatric indications. Neuropsychopharmacology 33, 685–700 (2008).

    Article  CAS  PubMed  Google Scholar 

  80. Kim, W. B. & Cho, J.-H. Encoding of contextual fear memory in hippocampal–amygdala circuit. Nat. Commun. 11, 1382 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institutes of Health (NIH) grant nos. NS087096, AG062904 and AG064580 (to C.D.W.), 1R01AR065538, 1R01CA193520, R01DK062472 and S10RR027931 (to R.J.S.), K01DK089145 and R01DK062472 (to A.B.S.); the Cure Alzheimer’s Fund (to C.D.W., S.H.C. and R.E.T.); an Alzheimer Association Research Grant (to C.D.W.), a SPARC Award from the McCance Center for Brain Health (to C.D.W.), the NeuroBehavior Laboratory Pilot Project Research Award from the Harvard NeuroDiscovery Center (to C.D.W.), the Hassenfeld Clinical Scholar Award (to C.D.W.), the Claflin Distinguished Scholar Award (to C.D.W.), the Harvard Brain Science Initiative Young Scientist Travel Award (to M.R.I.), the MSFHR (to B.R.C. and L.E.B.B.), the FRAXA (to B.R.C. and L.E.B.B.), the FXRFC (to B.R.C. and L.E.B.B.), the NSERC (to B.R.C. and L.E.B.B.), the CIHR (to B.R.C. and L.E.B.B.), the JPB Foundation (to B.M.S.) and the MGH Molecular Imaging Core (to R.J.S.). We thank H. Van Praag for critical comments on the study design, data analysis and the manuscript, and for providing us with the RV-CAG-GFP and RV-SYN-GTRgp. We acknowledge K. Gerber for technical assistance. We thank all members of the laboratory of C.D.W. for helpful discussions. We thank L. Djenoune for great help with Adobe Illustrator. We thank Z. Herbert from the Molecular Biology Core Facilities at the Dana-Farber Cancer Institute for support. We acknowledge J. Long for help with designing the AAV8-irisin. We acknowledge the MGH Viral Vector Core (supported by NIH/NINDS P30NS04776) and the Penn Vector core for technical and instrument support. Schematic icons in Figs. 4 and 5 were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

M.R.I., S.V., M.F.Y., R.L., E.B.H., S.F.B., S.M. and C.D.W. performed and analysed the in vivo experiments. S.V., E.B.H., M.R.I. and S.F.B. performed and analysed the in vitro experiments. M.P.J. provided conceptual and experiment advice on the irisin plasma analysis. R.R.K. performed bioinformatical analysis. L.E.B.B. and B.R.C. conceived, performed and analysed the electrophysiological experiments. A.B.S. and R.J.S. performed and analysed the dSTORM microscopy. H.T. and E.K. contributed to the data interpretation. H.K. and B.M.S. designed and generated AAV8-irisin with the Penn Vector core. B.J.C., S.H.C. and R.E.T. provided scientific input. B.M.S. provided conceptual advice. C.D.W. directed the research. M.R.I., S.V. and C.D.W. co-wrote the paper with assistance from all other authors.

Corresponding author

Correspondence to Christiane D. Wrann.

Ethics declarations

Competing interests

The authors declare the following competing interests: B.M.S. and C.D.W. hold a patent related to irisin (WO2015051007A1). B.M.S. and C.D.W. are academic co-founders and consultants for Aevum Therapeutics. C.D.W. has a financial interest in Aevum Therapeutics, a company developing drugs that harness the protective molecular mechanisms of exercise to treat neurodegenerative and neuromuscular disorders. C.D.W.’s interests were reviewed and are managed by MGH and Mass General Brigham in accordance with their conflict-of-interest policies. The other authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt. Nature Metabolism thanks the anonymous reviewers 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 Irisin deletion impairs cognitive function in exercise and aging.

a, Schematic of flox targeting-construct for the Fndc5 locus. b, Bodyweights (WT-sed n = 9; WT-run n = 12; F5KO-sed n = 12; F5KO-run, n = 14). c, qPCR of Fndc5 mRNA expression (n.d.: no detection) (n = 3 per group). d and e, Plasma irisin with commercial ELISA (n = 4 per group) (d) or EIA (n = 2 per group) (e). f, Rotarod, g, Grip strength, h, Gait scan analysis: average propulsion (left) and swing time (right). FR = front right limb, FL = front left, RR = rear right, RL = rear left (WT n = 6, F5KO n = 10), i, Open field test (OPF) (WT-sed n = 5; WT-run n = 6; F5KO-sed n = 6; F5KO-run n = 6), and j, Running activity and k and l, Morris-water-maze (MWM): latency to reach target platform (k) and 24 h probe trial in acquisition (l). NE (red bar) was the target quadrant (WT-sed n = 9; WT-run, n = 12; F5KO-sed n = 12; F5KO-run, n = 14). m, Swim speed, 9-months-old mice (WT, n = 10, F5KO, n = 10). n and o, Novel object recognition (NOR) task in young (n) (WT n = 12, F5KO, n = 14) and aged mice (o) (WT n = 5, F5KO, n = 7). p and q, Bodyweights (p) and Open field test (OPF) (q) for aged mice (WT n = 8, F5KO, n = 7). r, Spontaneous alternation behavior (SAB) in aged mice (WT, n = 7, F5KO, n = 7). s and t, Electrophysiology in DG using acute slices, paired pulse ratio (s) and EPSP input-output curve (t) (WT n = 14, F5KO n = 15 slices, 7 animals per group,). RM-Two-way ANOVA (k, t), Two-way ANOVA (b, h, i), One-way ANOVA. Significance was assigned only if time spent in the target quadrant was significantly different from all other quadrants (l), Two-tailed t-test (d-g, j, m-s). ***p < 0.001, ****p < 0.0001 n.s.= not significant. Data represented as mean ± SEM of biologically independent samples. See source data for exact p-values.

Source data

Extended Data Fig. 2 Pattern separation is impaired in F5KO mice and can be rescued by delivering irisin directly into the dentate gyrus.

a, Elevated plus maze. b, Tail suspension test. c and d, Baseline freezing in CFC-DL (c) and in CFC (d) of WT and F5KO mice. e and f, WT and F5KO stereotaxically injected with AAV8-GFP or AAV8-irisin-FLAG into the DG. Representative immunofluorescence images, GFP (green), irisin-FLAG (red), NeuN (magenta) in the hippocampus. Scale bar 200 µm (e) and baseline freezing in the CFC-DL (f) (WT n = 10, F5KO n = 12 (a, b, d); n = 9 per group (c); WT-GFP n = 10, WT-irisin n = 10, F5KO-GFP n = 9, F5KO-irisin n = 10 (f)). RM-Two-way ANOVA (b), One-Way ANOVA (f), Two-tailed t-test (a, c, d). n.s.= not significant. Data are represented as mean ± SEM of biologically independent samples.

Source data

Extended Data Fig. 3 Adult-born neurons in the hippocampus are altered in global F5KO mice.

a and b, Quantification (a) and representative immunohistochemistry images (b) of BrdU+ adult-born hippocampal neurons in WT and F5KO mice (n = 6 per group). Scale bar 100 μm. c and d, Quantification (c) and representative immunofluorescence images (d) of EdU+ adult-born hippocampal neurons in WT and F5KO mice with or without running exercise (WT-sed n = 8, WT-run n = 10, F5KO n = 8, F5KO-run n = 13). Scale bar 100 μm. e, Soma size of adult-born hippocampal neurons (WT-sed n = 60, WT-run n = 60, F5KO-sed n = 65, F5KO-run n = 64 neurons). f-h, Dendritic spine analysis of newborn neurons in the ventral hippocampus. Dendritic spines density (f), cumulative distribution of spine head width (g), and median spine head width (h) (WT-sed n = 7, WT-run n = 6, F5KO-sed n = 7, F5KO-run n = 6 (f); WT-sed n = 1239, WT-run n = 1056, F5KO-sed n = 1089, F5KO-run n = 1047 spines (g); WT-sed n = 7, WT-run n = 6, F5KO-sed n = 6, F5KO-run n = 6 (h)). i-l, Dendritic spine analysis in mature granule cells in the dentate gyrus of Thy1-GFP/WT and Thy1-GFP/F5KO. Representative confocal images of dendritic spines stained with anti-GFP (green). Scale bar 5 µm (i), dendritic spines density (j), cumulative distribution of spine head width (k), and median spine head width (l) (n = 5 per group (j, l); WT n = 921, F5KO n = 948 spines (k)). Two-way ANOVA (c, e, f, h), Kruskal-Wallis ANOVA (g), Kolmogorov-Smirnov test (k), Two-tailed t-test (a, j, l). *p < 0.05, ****p < 0.0001, n.s.= not significant. Data are represented as mean ± SEM of biologically independent samples, except for violin plots with center line = median, dotted line = upper and lower quartile (e). See source data for exact p-values.

Source data

Extended Data Fig. 4 The transcriptome of adult-born neurons in the hippocampus is altered in global F5KO mice.

a and b, Representative FACS density plots of single nuclei isolated from the hippocampus of a non-injected WT control mouse (a) and a RV-Syn-GTRgp-GFP injected mouse (b). Nuclei were stained with Vybrant DyeCycle stain to label intact nuclei. c, Volcano plot of DESeq2 analysis of bulk RNA-sequencing of microdissected dentate gyrus from F5KO and WT mice (n = 5 per group).

Extended Data Fig. 5 Genetic deletion of irisin impairs cognitive function in transgenic mouse models of AD.

a and b, MSD ELISA for Aβ-40 and Aβ-42 peptides in soluble fraction of hippocampus (a) and cortex (b) (APP/PS1-WT n = 2, APP/PS1-F5KO n = 3). c, Body weights at 6 months old. d and e, Open field test (OPF). f, Spontaneous Alternation Behavior (SAB). g, Baseline freezing in CFC. n = 10 per group (c, f, g); n = 7 per group (d, e). RM-Two-way ANOVA (d, e), Two-way ANOVA (a, b), Two-tailed t-test (c, f, g). **p < 0.01, n.s.= not significant. Data are represented as mean ± SEM of biologically independent samples. See source data for exact p-values.

Source data

Extended Data Fig. 6 Peripheral irisin improves cognitive function in APP/PS1 transgenic mouse model of AD.

APP/PS1 mice were injected with AAV8-GFP or AAV8-irisin-FLAG via the tail vein. a, Liver (GFP n = 14, irisin n = 11) b, Hippocampus “irisin” mRNA expression (GFP n = 5, irisin n = 5), c, Bodyweights at the beginning and end of the experiment, d and e, OPF test, f, SAB (GFP n = 15, irisin n = 11). g, Barnes Maze, escape latency to hole (GFP n = 9, irisin n = 5), h-j, Morris-water-maze (MWM) latency to reach the target platform (h) and 24 h probe trial in in acquisition (i). SW quadrant (blue bar) was the target quadrant. Latency to reach the target platform in reversal (j) (GFP n = 6, irisin n = 6). k and l, Baseline freezing (k) and freezing in CFC (l), m, CFC-DL (n = 6 per group). RM-Two-way ANOVA (c, d, e, g left, h, j, l, m), One-way ANOVA followed by Fisher’s LSD. Significance was assigned only if time spent in the target quadrant was significantly different from all other quadrants (i), Two-tailed t-test (b, f, g right, k), Two-tailed t-test with Welch’s correction (a). *p < 0.05, **p < 0.01, n.s.= not significant. Data are represented as mean ± SEM of biologically independent samples. See source data for exact p-values.

Source data

Extended Data Fig. 7 Peripheral irisin improves cognitive function in 5xFAD transgenic mouse model of AD.

5xFAD mice were injected with AAV8-GFP or AAV8-irisin-FLAG via the tail vein. a, Liver b, Hippocampus “irisin” mRNA expression (n = 3 per group). c, Bodyweights at the beginning and end of the experiment, d and e, OPF test, f, SAB. g-j, Morris-water-maze (MWM) latency to reach the target platform in acquisition (g) and 24 h probe trial in MWM in acquisition (h). NE quadrant (green bar) was the target quadrant. Latency to reach the target platform in reversal (i) and 24 h probe trial in MWM reversal (j). SW quadrant (green bar) was the target quadrant. k and l, CFC, baseline freezing (k) and freezing in CFC (l) (GFP n = 11, irisin n = 10 (c-f, k, l); GFP n = 8, irisin n = 7 (g-j)). RM-Two-way ANOVA (c, d, e, g, i, l), One-way ANOVA followed by Fisher’s LSD. Significance was assigned only if time spent in the target quadrant was significantly different from all other quadrants (h, j), Two-tailed t-test (a, b, f, k). *p < 0.05, **p < 0.01, ****p < 0.0001, n.s.= not significant, data are represented as mean ± SEM of biologically independent samples. See source data for exact p-values.

Source data

Extended Data Fig. 8 Peripheral irisin reduces glia activation in transgenic mouse models of AD.

APP/PS1 mice were injected with AAV8-GFP or AAV8-irisin-FLAG via the tail vein. a, Expression of synaptic plasticity genes in hippocampus derived from normalized read counts of RNA-seq analysis (GFP n = 5, irisin n = 5), b, MSD ELISA for Abeta40 peptide in cortex soluble fraction, c, MSD ELISA Abeta42 peptide in cortex soluble fraction (GFP n = 9, irisin n = 5). d and e, Representative immunofluorescence confocal images of hippocampus, GFAP (green), Iba-1 (red), 3D6 (Alexa 647). Scale bar 200 μm (d) and quantification of plaque burden in hippocampus (e) (GFP n = 7, irisin n = 5). f, Quantification of plaque burden cortex (GFP n = 5, irisin n = 3). g, Quantification of total BrdU+ cells (GFP n = 7, irisin n = 5). Two-way ANOVA (a), Two-tailed t-test (b, c, e-g). n.s.= not significant, data are represented as mean ± SEM of biologically independent samples.

Source data

Extended Data Fig. 9 Irisin binds αV/β5 integrin receptor on astrocytes in adult hippocampal neural stem cells cultures.

a, Two-color dSTORM images of integrin αV/β3 (left) and αV/β5 (right) (green) and irisin-FLAG (red) molecules. Scale bar 5 μm (irisin-FLAG-αV/β3 n = 7, irisin-FLAG-αV/β5 n = 10), b and c, Itgav gene expression (b) and Itgb5 gene expression (c) in the murine dentate gyrus from Linnarsson lab database (https://linnarssonlab.org/dentate/)59. d, QPCR analysis of mRNA expression of Map2, Dcx, Gfap, and Aif1 in the dentate gyrus (n = 6) and neurons differentiated from adult hippocampal stem cells (n = 8 from two independent experiments). Ct: cycle threshold value. Data are represented as mean ± SEM of biologically independent samples.

Source data

Extended Data Fig. 10 Peripherally delivered irisin has central effects.

a-e, WT mice injected with AAV8-GFP or AAV8-irisin-FLAG via the tail vein. Western blot of plasma from AAV8-GFP (1-2) and AAV8-irisin-FLAG (3-4) with or without deglycosylation (a), qPCR analysis of liver (b), quadriceps (c), inguinal white adipose tissue (iWAT) (d), interscapular brown adipose tissue (iBAT) (e). Two-way ANOVA (b-d). Data are represented as mean ± SEM of biologically independent samples (n = 6 per group) (b-e).

Source data

Supplementary information

Supplementary Information

Supplementary Methods, Supplementary Data 1–3, references and code

Reporting Summary

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Fig. 7

Numerical source data.

Source Data Fig. 8

Numerical source data.

Extended Data Fig. 1

Numerical source data.

Extended Data Fig. 2

Numerical source data.

Extended Data Fig. 3

Numerical source data.

Extended Data Fig. 5

Numerical source data.

Extended Data Fig. 6

Numerical source data.

Extended Data Fig. 7

Numerical source data.

Extended Data Fig. 8

Numerical source data.

Extended Data Fig. 9

Numerical source data.

Extended Data Fig. 10

Unmodified gel.

Extended Data Fig. 10

Numerical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Islam, M.R., Valaris, S., Young, M.F. et al. Exercise hormone irisin is a critical regulator of cognitive function. Nat Metab 3, 1058–1070 (2021). https://doi.org/10.1038/s42255-021-00438-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00438-z

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