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:

Dietary salt promotes cognitive impairment through tau phosphorylation

A Publisher Correction to this article was published on 14 January 2020

This article has been updated

Abstract

Dietary habits and vascular risk factors promote both Alzheimer’s disease and cognitive impairment caused by vascular factors1,2,3. Furthermore, accumulation of hyperphosphorylated tau, a microtubule-associated protein and a hallmark of Alzheimer’s pathology4, is also linked to vascular cognitive impairment5,6. In mice, a salt-rich diet leads to cognitive dysfunction associated with a nitric oxide deficit in cerebral endothelial cells and cerebral hypoperfusion7. Here we report that dietary salt induces hyperphosphorylation of tau followed by cognitive dysfunction in mice, and that these effects are prevented by restoring endothelial nitric oxide production. The nitric oxide deficiency reduces neuronal calpain nitrosylation and results in enzyme activation, which, in turn, leads to tau phosphorylation by activating cyclin-dependent kinase 5. Salt-induced cognitive impairment is not observed in tau-null mice or in mice treated with anti-tau antibodies, despite persistent cerebral hypoperfusion and neurovascular dysfunction. These findings identify a causal link between dietary salt, endothelial dysfunction and tau pathology, independent of haemodynamic insufficiency. Avoidance of excessive salt intake and maintenance of vascular health may help to stave off the vascular and neurodegenerative pathologies that underlie dementia in the elderly.

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: HSD increases tau phosphorylation and insoluble tau.
Fig. 2: The NO precursor l-arginine prevents the increase in p-tau induced by HSD.
Fig. 3: HSD induces activation of calpain and CDK5 associated with calpain denitrosylation.
Fig. 4: HSD-induced cognitive dysfunction is not observed in tau−/− mice and is prevented by tau antibodies despite cerebrovascular insufficiency.

Similar content being viewed by others

Data availability

Source data include final quantifications from in vivo animal work.

Change history

  • 14 January 2020

    An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Scarmeas, N., Anastasiou, C. A. & Yannakoulia, M. Nutrition and prevention of cognitive impairment. Lancet Neurol. 17, 1006–1015 (2018).

    Article  PubMed  Google Scholar 

  2. van de Rest, O., Berendsen, A. A., Haveman-Nies, A. & de Groot, L. C. Dietary patterns, cognitive decline, and dementia: a systematic review. Adv. Nutr. 6, 154–168 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Kendig, M. D. & Morris, M. J. Reviewing the effects of dietary salt on cognition: mechanisms and future directions. Asia Pac. J. Clin. Nutr. 28, 6–14 (2019).

    PubMed  Google Scholar 

  4. De Strooper, B. & Karran, E. The cellular phase of Alzheimer’s disease. Cell 164, 603–615 (2016).

    Article  PubMed  Google Scholar 

  5. Nation, D. A. et al. Pulse pressure in relation to tau-mediated neurodegeneration, cerebral amyloidosis, and progression to dementia in very old adults. JAMA Neurol. 72, 546–553 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Kim, H. J. et al. Assessment of extent and role of tau in subcortical vascular cognitive impairment using 18F-AV1451 positron emission tomography imaging. JAMA Neurol. 75, 999–1007 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Faraco, G. et al. Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response. Nat. Neurosci. 21, 240–249 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fiocco, A. J. et al. Sodium intake and physical activity impact cognitive maintenance in older adults: the NuAge Study. Neurobiol. Aging 33, 829.e821–829.e28, (2012).

    Article  CAS  Google Scholar 

  9. Gardener, H., Rundek, T., Wright, C. B., Elkind, M. S. & Sacco, R. L. Dietary sodium and risk of stroke in the Northern Manhattan study. Stroke 43, 1200–1205 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Blumenthal, J. A. et al. Lifestyle and neurocognition in older adults with cognitive impairments: a randomized trial. Neurology 92, e212–e223 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Heye, A. K. et al. Blood pressure and sodium: association with MRI markers in cerebral small vessel disease. J. Cereb. Blood Flow Metab. 36, 264–274 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Iadecola, C. The pathobiology of vascular dementia. Neuron 80, 844–866 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Sweeney, M. D. et al. Vascular dysfunction—the disregarded partner of Alzheimer’s disease. Alzheimers Dement. 15, 158–167 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Shi, Y. et al. Cerebral blood flow in small vessel disease: a systematic review and meta-analysis. J. Cereb. Blood Flow Metab. 36, 1653–1667 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Marshall, R. S. et al. Recovery of brain function during induced cerebral hypoperfusion. Brain 124, 1208–1217 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Wang, Y. & Mandelkow, E. Tau in physiology and pathology. Nat. Rev. Neurosci. 17, 5–21 (2016).

    Article  PubMed  CAS  Google Scholar 

  17. Zhao, Y. et al. Sodium intake regulates glucose homeostasis through the PPARδ/adiponectin-mediated SGLT2 pathway. Cell Metab. 23, 699–711 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Min, S. W. et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat. Med. 21, 1154–1162 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Iadecola, C. et al. SOD1 rescues cerebral endothelial dysfunction in mice overexpressing amyloid precursor protein. Nat. Neurosci. 2, 157–161 (1999).

    Article  CAS  PubMed  Google Scholar 

  20. Faraco, G. et al. Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension. J. Clin. Invest. 126, 4674–4689 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Arendt, T., Stieler, J. T. & Holzer, M. Tau and tauopathies. Brain Res. Bull. 126, 238–292 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Lee, M. S. et al. Neurotoxicity induces cleavage of p35 to p25 by calpain. Nature 405, 360–364 (2000).

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Patrick, G. N. et al. Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration. Nature 402, 615–622 (1999).

    Article  ADS  CAS  PubMed  Google Scholar 

  24. Austin, S. A. & Katusic, Z. S. Loss of endothelial nitric oxide synthase promotes p25 generation and tau phosphorylation in a murine model of Alzheimer’s disease. Circ. Res. 119, 1128–1134 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bibb, J. A. et al. Phosphorylation of DARPP-32 by Cdk5 modulates dopamine signalling in neurons. Nature 402, 669–671 (1999).

    Article  ADS  CAS  PubMed  Google Scholar 

  26. Shukla, V. et al. A truncated peptide from p35, a Cdk5 activator, prevents Alzheimer’s disease phenotypes in model mice. FASEB J. 27, 174–186 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kimura, T. et al. Isomerase Pin1 stimulates dephosphorylation of tau protein at cyclin-dependent kinase (Cdk5)-dependent Alzheimer phosphorylation sites. J. Biol. Chem. 288, 7968–7977 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ono, Y., Saido, T. C. & Sorimachi, H. Calpain research for drug discovery: challenges and potential. Nat. Rev. Drug Discov. 15, 854–876 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Etwebi, Z., Landesberg, G., Preston, K., Eguchi, S. & Scalia, R. Mechanistic role of the calcium-dependent protease calpain in the endothelial dysfunction induced by MPO (myeloperoxidase). Hypertension 71, 761–770 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Qu, J. et al. S-Nitrosylation activates Cdk5 and contributes to synaptic spine loss induced by β-amyloid peptide. Proc. Natl Acad. Sci. USA 108, 14330–14335 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Iadecola, C. The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron 96, 17–42 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yanamandra, K. et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 80, 402–414 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Powles, J. et al. Global, regional and national sodium intakes in 1990 and 2010: a systematic analysis of 24 h urinary sodium excretion and dietary surveys worldwide. BMJ Open 3, e003733 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Hochrainer, K. et al. The ubiquitin ligase HERC3 attenuates NF-κB-dependent transcription independently of its enzymatic activity by delivering the RelA subunit for degradation. Nucleic Acids Res. 43, 9889–9904 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Shukla, V. et al. TFP5, a peptide inhibitor of aberrant and hyperactive Cdk5/p25, attenuates pathological phenotypes and restores synaptic function in CK-p25Tg mice. J. Alzheimers Dis. 56, 335–349 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Faraco, G. et al. Circulating endothelin-1 alters critical mechanisms regulating cerebral microcirculation. Hypertension 62, 759–766 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Kober, F. et al. High-resolution myocardial perfusion mapping in small animals in vivo by spin-labeling gradient-echo imaging. Magn. Reson. Med. 51, 62–67 (2004).

    Article  PubMed  Google Scholar 

  38. Petry, F. R. et al. Specificity of anti-tau antibodies when analyzing mice models of Alzheimer’s disease: problems and solutions. PLoS One 9, e94251 (2014).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  39. Faraco, G. et al. Hypertension enhances Aβ-induced neurovascular dysfunction, promotes β-secretase activity, and leads to amyloidogenic processing of APP. J. Cereb. Blood Flow Metab. 36, 241–252 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Voit, A. et al. Reducing sarcolipin expression mitigates Duchenne muscular dystrophy and associated cardiomyopathy in mice. Nat. Commun. 8, 1068 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  41. Liu, W. et al. Metabolic stress-induced cardiomyopathy is caused by mitochondrial dysfunction due to attenuated Erk5 signaling. Nat. Commun. 8, 494 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  42. Forrester, M. T., Foster, M. W., Benhar, M. & Stamler, J. S. Detection of protein S-nitrosylation with the biotin-switch technique. Free Radic. Biol. Med. 46, 119–126 (2009).

    Article  CAS  PubMed  Google Scholar 

  43. Cohen, S. J. & Stackman, R. W. Jr Assessing rodent hippocampal involvement in the novel object recognition task. A review. Behav. Brain Res. 285, 105–117 (2015).

    Article  PubMed  Google Scholar 

  44. Grayson, B. et al. Assessment of disease-related cognitive impairments using the novel object recognition (NOR) task in rodents. Behav. Brain Res. 285, 176–193 (2015).

    Article  PubMed  Google Scholar 

  45. O’Leary, T. P. & Brown, R. E. Optimization of apparatus design and behavioral measures for the assessment of visuo-spatial learning and memory of mice on the Barnes maze. Learn. Mem. 20, 85–96 (2013).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank P. Davies for providing the RZ3, MC1 and PHF1 antibodies and Y. Li for sharing the Quickplex SQ 120 system (Meso Scale Diagnostics LLC). This study was supported by National Institutes of Health grants R37-NS089323 (C.I.) and 1R01-NS095441 (C.I.), by a grant from the Cure Alzheimer’s Fund (G.F. and C.I.) and by a Scientist Development Grant from the American Heart Association (G.F.). Support from the Feil Family Foundation is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

G.F. performed western blotting experiments, behavioural tests and cerebrovascular studies, and analysed data. K.H. performed experiments on CDK5 and GSK3β activity and analysed data. S.G.S. performed western blotting experiments, behavioural tests and immunohistochemistry. S.S. and M.M.S. performed experiments on the effects of hypertension on tau. A.M. performed immunohistochemistry experiments. H.J. and D.M.H. provided the HJ8.8 antibody. J.A. supervised the molecular aspects of the study and edited the manuscript. G.F. and C.I. designed and supervised the entire study and wrote the manuscript.

Corresponding authors

Correspondence to Giuseppe Faraco or Costantino Iadecola.

Ethics declarations

Competing interests

D.M.H. is listed as an inventor on a patent licensed by Washington University to C2N Diagnostics and subsequently AbbVie on the therapeutic use of anti-tau antibodies; co-founded and is on the scientific advisory board of C2N Diagnostics; and is on the scientific advisory board of Denali, Genentech, and Proclara. C.I. is on the scientific advisory board of Broadview Ventures.

Additional information

Peer review information Nature thanks Nikolaos Scarmeas, Berislav Zlokovic 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 figures and tables

Extended Data Fig. 1 HSD (4 or 8%) induced tau phosphorylation: brain localization, sex differences and time course.

a, HSD (8% NaCl) increases tau phosphorylation on Ser396 (PHF13) and on Ser199Ser202 in the hippocampus (HIPP) but not in the neocortex (CTX) (HIPP: PHF13, ND/HSD n = 4/5, *P = 0.0016; Ser199Ser202, ND/HSD n = 4/5, *P = 0.0337; two-tailed unpaired t-test versus ND), whereas acetylation of tau on Lys280 (K280) is not affected. MC1 immunoreactivity increases in both neocortex and hippocampus in HSD-fed mice but reaches statistical significance only in neocortex (MC1, ND/HSD n = 4/5, *P = 0.0321 versus ND, two-tailed unpaired t-test). b, Tau phosphorylation on Ser199Ser202 and Ser202Thr205 is abolished after treatment of brain samples with lambda phosphatase. c, HSD increases AT8 immunoreactivity (left graph) in neocortex and hippocampus of female mice, but RZ3 (middle) increases only in neocortex (AT8, cortex: ND/HSD n = 8/10, *P = 0.0159; hippocampus: ND/HSD n = 10/9, *P = 0.0151; RZ3, cortex: ND/HSD n = 10/8, *P = 0.0117; two-tailed unpaired t-test for HSD versus ND). Right, HSD induces a deficit in NOR in female mice (ND/HSD n = 8/9, *P = 0.0017 versus ND, two-tailed unpaired t-test). d, HSD increases AT8 immunoreactivity in neuronal cell bodies of the somatosensory cortex (scale bars, 100 μm (main images); 10 µm (insets)) and MC1 immunoreactivity in neuronal bodies of the pyriform cortex (scale bars, 500 μm (main images); 100 µm (insets)). Representative images from ND- and HSD-fed mice (n = 5 per group). e, Thioflavin S staining is not present in mice fed an HSD, indicating absence of neurofibrillary tangles, which can be observed in rTg4510 mice (scale bars, 500 μm (main images); 100 µm (inset)). Representative images from n = 5 ND- and HSD-fed mice and n = 3 rTg4510 mice. f, HSD (4%) increases AT8 immunoreactivity in the neocortex but not in the hippocampus (AT8, cortex: ND/HSD n = 5/5, *P = 0.0148 versus ND, two-tailed unpaired t-test). RZ3 was not increased in both regions. g, Time course of the increase in AT8 and RZ3 induced by HSD in the hippocampus. AT8 levels are increased after 4 weeks of HSD. RZ3 levels are increased after 4, 12, 24 and 36 weeks of HSD (AT8, 4 weeks: ND/HSD n = 4/5, *P = 0.0386; 12 weeks: ND/HSD n = 9/9, *P < 0.0001; RZ3, 4 weeks: ND/HSD n = 4/5, *P = 0.0041; RZ3, 12 weeks: ND/HSD n = 9/9, *P = 0.0011; 24 weeks: ND/HSD n = 7/10, *P = 0.0017; 36 weeks: ND/HSD n = 5/4, *P = 0.0188; two-tailed unpaired t-test for HSD versus ND). For gel source data see Supplementary Fig. 1. Data are expressed as mean ± s.e.m.

Source data

Extended Data Fig. 2 Effect of HSD on neurons, astrocytes, microglia/macrophages, pericytes and white-matter integrity.

a, HSD (NaCl 8%) does not affect neurons (NEUN), astrocytes (GFAP), microglia/macrophages (IBA-1) (cortex: microglia, ND/HSD n = 5/5, P = 0.0570; hippocampus: ND/HSD n = 5/5, P = 0.0556; two-tailed unpaired t-test for HSD versus ND) or pericytes (CD13) in both the pyriform cortex and the hippocampus (scale bars, 500 μm (except where specified)). b, No evidence of neuronal cell death is observed in HSD-fed mice by Fluoro-Jade B or TUNEL staining (scale bar, 500 μm). +DNase indicates positive control for TUNEL staining. Representative images from ND- and HSD-fed mice (n = 5 per group). c, Klüver–Barrera stain shows no white-matter damage in the corpus callosum of HSD-fed mice (scale bar, 100 μm). Representative images from ND- and HSD-fed mice (n = 4 per group). Data are expressed as mean ± s.e.m.

Source data

Extended Data Fig. 3 Aβ levels in HSD-fed mice and correlation of behavioural deficits with p-tau, as well as p-tau in hypertension, HSD-treated tg2576 mice and hypothermia.

a, HSD (8% NaCl) does not alter the distance travelled before finding the escape hole in the Barnes maze (primary distance, ND/HSD n = 13/13, diet: *P = 0.0462, time: *P < 0.0001, two-way repeated measures ANOVA and Bonferroni’s test; primary distance day 5: ND/HSD n = 13/13, P = 0.0670 versus ND, two-tailed unpaired t-test) or the number of errors made (primary errors, ND/HSD n = 13/13, diet: P = 0.110, time: *P = 0.0004, two-way repeated measures ANOVA and Bonferroni’s test; primary errors day 5: P = 0.1226 versus ND, two-tailed unpaired t-test). b, RZ3 levels in the cortex correlate with cognitive performance on the NOR test. No correlation was found between hippocampal RZ3 levels and cognitive performance on either the Barnes maze or the NOR test. RZ3 cortex: Barnes maze r = 0.2828, *P = 0.0133, n = 76; NOR r = −0.2806, *P = 0.0170, n = 72; RZ3 hippocampus: Barnes maze r = 0.1739, P = 0.1470, n = 71; NOR r = −0.1746, P = 0.1577, n = 67, Pearson’s correlation coefficient). c, HSD does not increase soluble or insoluble Aβ38, Aβ40 or Aβ42 in the neocortex. Aβ38, soluble ND/HSD n = 11/9, insoluble ND/HSD n = 7/6; Aβ40, soluble ND/HSD n = 11/14, insoluble ND/HSD n = 7/6; Aβ42, soluble ND/HSD n = 9/9, insoluble ND/HSD n = 7/6. d, Delivery of angiotensin II (ANGII; 600 ng kg−1 min−1, subcutaneously (s.c.)) via osmotic minipumps over 6 weeks increases systolic blood pressure (SBP) and induces cognitive deficits (SBP: Veh/ANGII n = 10/10, treatment: *P < 0.0001, time: *P < 0.0001, repeated measures two-way ANOVA and Bonferroni’s test; NOR: 2 weeks Veh/ANGII n = 12/12, 4 weeks Veh/ANGII n = 10/11, 6 weeks Veh/ANGII n = 7/7, treatment: *P < 0.0021, time: *P = 0.0208, two-way ANOVA and Bonferroni’s test). e, Administration of angiotensin II increases AT8 and RZ3 immunoreactivity in the neocortex but not the hippocampus (cortex, AT8 6 weeks: Veh/ANGII n = 4/4, *P = 0.0324; RZ3 6 weeks: Veh/ANGII n = 5/5, *P = 0.0262; hippocampus, AT8 6 weeks: Veh/ANGII n = 5/5, P = 0.4056; RZ3 6 weeks: Veh/ANGII n = 5/5, P = 0.0556, two-tailed unpaired t-test versus vehicle). f, HSD increases AT8 and RZ3 levels in both the neocortex and the hippocampus of 6-month-old Tg2576 mice (cortex, AT8: *P < 0.0001; hippocampus, AT8: *P = 0.0153; cortex, RZ3: *P < 0.0001; hippocampus, RZ3: *P = 0.0239; two-tailed unpaired t-test for HSD versus ND). g, Hypothermia induces massive AT8 phosphorylation (cortex: AT8 n = 4/5, *P = 0.0159; hippocampus: AT8 n = 4/5, *P = 0.0159) and increases MC1 (cortex: MC1 n = 4/5, *P = 0.0317; hippocampus: MC1 n = 4/5, *P = 0.0159) and RZ3 (cortex: RZ3 n = 4/5, *P = 0.0201; hippocampus: RZ3 n = 4/5, *P = 0.0453). Unpaired two-tailed t-test for hypothermia (HYPO) versus normal temperature (NT). h, Unlike HSD (Fig. 1G), hypothermia does not shift tau from soluble to more insoluble fractions. For gel source data see Supplementary Fig. 1. Data are expressed as mean ± s.e.m.

Source data

Extended Data Fig. 4 Effect of l-arginine on p-tau and calpain expression, as well as p-tau in eNOS−/− mice, calpain and CDK5 localization, pDARPP-32 with HSD, and IL-17 levels.

a, Administration of l-arginine (10 g l−1 in drinking water), starting at week 8 of HSD and continuing through week 12, suppresses RZ3 levels in the neocortex but not in the hippocampus (cortex: RZ3, ND/HSD n = 10/10, *P < 0.0001; hippocampus: RZ3, ND/HSD n = 10/10, *P = 0.0005, two-tailed unpaired t-test versus normal diet with vehicle). b, l-Arginine does not affect the increase in serum IL-17 induced by HSD (Veh, ND/HSD n = 9/11, *P = 0.0002 versus ND Veh; l-arg, ND/HSD n = 9/8, *P < 0.0001 versus ND l-arg, two-tailed unpaired t-test). c, AT8 and RZ3 levels are elevated in the neocortex and hippocampus of eNOS−/− mice on ND (AT8: cortex, ND/HSD n = 5/4, *P = 0.0029; hippocampus, ND/HSD n = 5/4, *P = 0.0078; RZ3: cortex, ND/HSD n = 5/4, *P = 0.0003; hippocampus, ND/HSD n = 5/4, *P = 0.0128, two-tailed unpaired t-test versus wild-type mice). d, HSD does not increase tau phosphorylation in eNOS−/− mice (RZ3: hippocampus, ND/HSD n = 7/8, *P = 0.0224 versus ND, two-tailed unpaired t-test). e, Calpain 2 immunoreactivity is present in neuronal cell bodies of the somatosensory and piriform cortex (scale bars, 500 μm (left); 100 µm (right)). Representative images from n = 3 mice. f, Colocalization of Calpain 2 and CDK5 in neuronal cell bodies of the piriform cortex (scale bars, 50 μm (main images); 10 µm (inset)). Representative images from n = 3 mice. g, HSD has no effect on the phosphorylation of the CDK5 substrate DARPP-32 in neocortex; ND/HSD n = 10/10. h, Administration of the CDK5 peptide inhibitor TFP5 has no effect on the increase in serum IL-17 levels induced by HSD (scrambled: ND/HSD n = 5/4, *P = 0.0002 versus ND scrambled; TFP5: ND/HSD n = 7/8, *P < 0.359 versus ND TFP5; two-tailed unpaired t-test). i, l-Arginine does not alter the levels of calpain 1 and 2 in the neocortex or hippocampus. ND/HSD n = 3/5. For gel source data see Supplementary Fig. 1. Data are expressed as mean ± s.e.m.

Source data

Extended Data Fig. 5 GSK3β, PIN-1, calpastatin and CDK5 nitrosylation in HSD-fed mice, as well as neurovascular coupling, effect of HJ8.8 on p-tau, serum IL-17, and summary.

a, HSD has no effect on the expression or activity of GSK3β in the neocortex. ND/HSD n = 10/10. b, HSD does not alter the expression of the prolyl cis/trans isomerase PIN-1, a regulator of tau dephosphorylation. ND/HSD n = 5/5. c, The expression of calpastatin, an endogenous inhibitor of calpain activity, is not reduced by HSD. ND/HSD n = 10/10. d, Nitrosylation of CDK5 is reduced in the neocortex of HSD-fed mice (ND/HSD n = 9/9, diet: *P = 0.0143; ascorbate: *P < 0.0001; two-way ANOVA and Tukey’s test). e, HJ8.8 reduces AT8 in the hippocampus (IgG: ND/HSD n = 13/12; HJ8.8: ND/HSD n = 9/13; *P < 0.0001, Kruskal–Wallis test and Dunn’s test). RZ3 levels are not altered by HJ8.8. f, Administration of HJ8.8 does alter the increase in serum IL-17 levels induced by HSD (IgG: ND/HSD n = 9/9, *P = 0.0192 versus ND IgG; HJ8.8: ND/HSD n = 7/5, *P = 0.0421 versus ND HJ8.8, two-tailed unpaired t-test). g, The increase in somatosensory cortex CBF induced by neural activity evoked by mechanical stimulation of the whiskers is not reduced by HSD in wild-type, tau−/− or HJ8.8-treated mice (wild-type ND/HSD n = 5/7, tau−/− ND/HSD n = 9/8; IgG ND/HSD n = 5/5, HJ8.8 ND/HSD n = 5/5). h, Western blotting showing enrichment of tau in boiled RIPA neocortical samples (heat-stable fraction, HS). Note that β-actin is lost during the boiling process. Representative images from n = 3 experiments. i, Cartoon depicting the mechanisms by which HSD leads to tau phosphorylation and cognitive impairment. HSD elicits a TH17 response in the small intestine, which leads to an increase in circulating IL-17. IL-17, in turn, suppresses endothelial NO production by inducing inhibitory phosphorylation of eNOS at Thr495. The NO deficit results in reduced nitrosylation of calpain in neurons, and increases in calpain activity, p35 to p25 cleavage, activation of CDK5, and tau phosphorylation, which is ultimately responsible for cognitive dysfunction. In support of this chain of events, rescuing the endothelial NO deficit with l-arginine, lack of tau in tau-null mice, treatment with the CDK5 peptide inhibitor TFP5 or treatment with antibodies directed against tau (Tau ab) prevent the cognitive dysfunction. For gel source data see Supplementary Fig. 1. Data are expressed as mean ± s.e.m.

Source data

Supplementary information

Supplementary Figures

This file contains full immunoblots images for the figures and extended figures.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Faraco, G., Hochrainer, K., Segarra, S.G. et al. Dietary salt promotes cognitive impairment through tau phosphorylation. Nature 574, 686–690 (2019). https://doi.org/10.1038/s41586-019-1688-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1688-z

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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