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.

Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response

Abstract

A diet rich in salt is linked to an increased risk of cerebrovascular diseases and dementia, but it remains unclear how dietary salt harms the brain. We report that, in mice, excess dietary salt suppresses resting cerebral blood flow and endothelial function, leading to cognitive impairment. The effect depends on expansion of TH17 cells in the small intestine, resulting in a marked increase in plasma interleukin-17 (IL-17). Circulating IL-17, in turn, promotes endothelial dysfunction and cognitive impairment by the Rho kinase–dependent inhibitory phosphorylation of endothelial nitric oxide synthase and reduced nitric oxide production in cerebral endothelial cells. The findings reveal a new gut–brain axis linking dietary habits to cognitive impairment through a gut-initiated adaptive immune response compromising brain function via circulating IL-17. Thus, the TH17 cell–IL-17 pathway is a putative target to counter the deleterious brain effects induced by dietary salt and other diseases associated with TH17 polarization.

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: HSD reduces resting CBF and induces endothelial dysfunction, effects reversed by returning to a normal diet.
Fig. 2: HSD induces delayed cognitive dysfunction.
Fig. 3: The NO precursor l-arginine reverses the neurovascular and cognitive dysfunction of HSD.
Fig. 4: HSD increases inhibitory eNOS phosphorylation.
Fig. 5: HSD induces TH17 differentiation in the small intestine and increases IL-17 plasma levels.
Fig. 6: The neurovascular and cognitive effects of HSD are not observed in mice lacking IL-17 or lymphocytes (Rag1−/− mice).
Fig. 7: The neurovascular and the cognitive effects of HSD are prevented by IL-17-neutralizing antibodies and reproduced by IL-17 administration in mice fed a normal diet.
Fig. 8: IL-17 suppresses NO production via ROCK, and ROCK inhibition ameliorates the neurovascular and cognitive dysfunction of HSD.

Similar content being viewed by others

References

  1. Mozaffarian, D. et al. Global sodium consumption and death from cardiovascular causes. N. Engl. J. Med. 371, 624–634 (2014).

    Article  PubMed  Google Scholar 

  2. Zemel, M. B. & Sowers, J. R. Salt sensitivity and systemic hypertension in the elderly. Am. J. Cardiol. 61, 7H–12H (1988).

    Article  CAS  PubMed  Google Scholar 

  3. Farquhar, W. B., Edwards, D. G., Jurkovitz, C. T. & Weintraub, W. S. Dietary sodium and health: more than just blood pressure. J. Am. Coll. Cardiol. 65, 1042–1050 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Appel, L. J. et al. The importance of population-wide sodium reduction as a means to prevent cardiovascular disease and stroke: a call to action from the American Heart Association. Circulation 123, 1138–1143 (2011).

    Article  PubMed  Google Scholar 

  5. Institute of Medicine (US) Committee on Strategies to Reduce Sodium Intake. Strategies to Reduce Sodium Intake in the United States. (Henney, J.E., Taylor, C.L. & Boon, C.S. eds.) (National Academies Press, Washington DC, 2010).

  6. Kotchen, T. A., Cowley, A. W. Jr. & Frohlich, E. D. Salt in health and disease—a delicate balance. N. Engl. J. Med. 368, 1229–1237 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Nicholls, M. G. Population-wide dietary sodium restriction: a cautious view. Curr. Hypertens. Rep. 13, 325–327 (2011).

    Article  PubMed  Google Scholar 

  8. Oh, Y. S. et al. National Heart, Lung, and Blood Institute Working Group report on salt in human health and sickness: building on the current scientific evidence. Hypertension 68, 281–288 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Strazzullo, P., D’Elia, L., Kandala, N.-B. & Cappuccio, F. P. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. Br. Med. J. 339, b4567 (2009).

    Article  Google Scholar 

  11. Boegehold, M. A. The effect of high salt intake on endothelial function: reduced vascular nitric oxide in the absence of hypertension. J. Vasc. Res. 50, 458–467 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Cosic, A. et al. Attenuated flow-induced dilatation of middle cerebral arteries is related to increased vascular oxidative stress in rats on a short-term high salt diet. J. Physiol. (Lond.) 594, 4917–4931 (2016).

    Article  CAS  Google Scholar 

  13. 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  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Kleinewietfeld, M. et al. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature 496, 518–522 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wu, C. et al. Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature 496, 513–517 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kebir, H. et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat. Med. 13, 1173–1175 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nguyen, H. et al. Interleukin-17 causes Rho-kinase-mediated endothelial dysfunction and hypertension. Cardiovasc. Res. 97, 696–704 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. Cochet, H. et al. Comprehensive phenotyping of salt-induced hypertensive heart disease in living mice using cardiac magnetic resonance. Eur. Radiol. 23, 332–338 (2013).

    Article  PubMed  Google Scholar 

  21. Schlote, J. et al. Cardiovascular and renal effects of high salt diet in GDNF+/- mice with low nephron number. Kidney Blood Press. Res. 37, 379–391 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Jackman, K. A. et al. Dichotomous effects of chronic intermittent hypoxia on focal cerebral ischemic injury. Stroke 45, 1460–1467 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Toda, N., Ayajiki, K. & Okamura, T. Cerebral blood flow regulation by nitric oxide: recent advances. Pharmacol. Rev. 61, 62–97 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. Wang, G. et al. Angiotensin II slow-pressor hypertension enhances NMDA currents and NOX2-dependent superoxide production in hypothalamic paraventricular neurons. Am. J. Physiol. Regul. Integr. Comp. Physiol. 304, R1096–R1106 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chen, B. R., Kozberg, M. G., Bouchard, M. B., Shaik, M. A. & Hillman, E. M. C. A critical role for the vascular endothelium in functional neurovascular coupling in the brain. J. Am. Heart Assoc. 3, e000787 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Poggesi, A., Pasi, M., Pescini, F., Pantoni, L. & Inzitari, D. Circulating biologic markers of endothelial dysfunction in cerebral small vessel disease: a review. J. Cereb. Blood Flow. Metab. 36, 72–94 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cohen, S. J. et al. The rodent hippocampus is essential for nonspatial object memory. Curr. Biol. 23, 1685–1690 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Barnes, C. A. Memory deficits associated with senescence: a neurophysiological and behavioral study in the rat. J. Comp. Physiol. Psychol. 93, 74–104 (1979).

    Article  CAS  PubMed  Google Scholar 

  29. Deacon, R. M. Assessing nest building in mice. Nat. Protoc. 1, 1117–1119 (2006).

    Article  PubMed  Google Scholar 

  30. Rosenblum, W. I., Nelson, G. H. & Shimizu, T. L-Arginine suffusion restores response to acetylcholine in brain arterioles with damaged endothelium. Am. J. Physiol. 262, H961–H964 (1992).

    CAS  PubMed  Google Scholar 

  31. Harris, M. B. et al. Reciprocal phosphorylation and regulation of endothelial nitric-oxide synthase in response to bradykinin stimulation. J. Biol. Chem. 276, 16587–16591 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Smith, L., Payne, J. A., Sedeek, M. H., Granger, J. P. & Khalil, R. A. Endothelin-induced increases in Ca2+ entry mechanisms of vascular contraction are enhanced during high-salt diet. Hypertension 41, 787–793 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  34. Benakis, C. et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat. Med. 22, 516–523 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hernandez, A. L. et al. Sodium chloride inhibits the suppressive function of FOXP3+ regulatory T cells. J. Clin. Invest. 125, 4212–4222 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Kipnis, J. Multifaceted interactions between adaptive immunity and the central nervous system. Science 353, 766–771 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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 

  38. Binger, K. J. et al. High salt reduces the activation of IL-4- and IL-13-stimulated macrophages. J. Clin. Invest. 125, 4223–4238 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Mandala, S. et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 296, 346–349 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Sugimoto, M. et al. Rho-kinase phosphorylates eNOS at threonine 495 in endothelial cells. Biochem. Biophys. Res. Commun. 361, 462–467 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Guterbaum, T. J. et al. Endothelial nitric oxide synthase phosphorylation at Threonine 495 and mitochondrial reactive oxygen species formation in response to a high H2O2 concentration. J. Vasc. Res. 50, 410–420 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Katusic, Z. S. & Austin, S. A. Endothelial nitric oxide: protector of a healthy mind. Eur. Heart J. 35, 888–894 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Love, S. & Miners, J. S. Cerebral hypoperfusion and the energy deficit in Alzheimer’s disease. Brain Pathol. 26, 607–617 (2016).

    Article  PubMed  Google Scholar 

  44. Garthwaite, J. Concepts of neural nitric oxide-mediated transmission. Eur. J. Neurosci. 27, 2783–2802 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Gu, C., Wu, L. & Li, X. IL-17 family: cytokines, receptors and signaling. Cytokine 64, 477–485 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Tablazon, I. L. D., Al-Dabagh, A., Davis, S. A. & Feldman, S. R. Risk of cardiovascular disorders in psoriasis patients: current and future. Am. J. Clin. Dermatol. 14, 1–7 (2013).

    Article  PubMed  Google Scholar 

  47. Liou, T.-H. et al. Risk of stroke in patients with rheumatism: a nationwide longitudinal population-based study. Sci. Rep. 4, 5110 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ferro, J. M., Oliveira, S. N. & Correia, L. Neurologic manifestations of inflammatory bowel diseases. Handb. Clin. Neurol. 120, 595–605 (2014).

    Article  PubMed  Google Scholar 

  49. Marshall, O. et al. Impaired cerebrovascular reactivity in multiple sclerosis. JAMA Neurol. 71, 1275–1281 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Park, L. et al. Scavenger receptor CD36 is essential for the cerebrovascular oxidative stress and neurovascular dysfunction induced by amyloid-beta. Proc. Natl. Acad. Sci. USA 108, 5063–5068 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge support from the Feil Family Foundation. This work was supported by National Institutes of Health grants R37-NS089323 (C.I.) and 1R01-NS095441 (C.I.), by a Scientist Development Grant from the American Heart Association (G.F.) and by a network grant from the Fondation Leducq (Sphingonet) (C.I., J.A., G.F.).

Author information

Authors and Affiliations

Authors

Contributions

Study design: C.I., J.A. and G.F. Conducting experiments and acquiring data: G.F., D.B., L.G.B., G.W., G.R., H.C., I.B., M.M.S., S.G.S., K.K., Y.S., M.M. and H.V. Analyzing data: G.F., D.B., G.W., H.C., I.B., M.M.S. and K.K. Writing the manuscript: G.F. and C.I.

Corresponding author

Correspondence to Costantino Iadecola.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Effect of HSD on body weight, energy intake, systolic blood pressure, hippocampal resting CBF and CBF response to adenosine and whisker stimulation.

A: HSD tends to reduce body weight and increase caloric intake, but the effect is not statistically significant; Diet: p = 0.2668, Time: p < 0.0001; ND/HSD n = 15/20 mice/group (Two-way ANOVA and Tukey’s test). B: HSD does not alter systolic blood pressure measured in awake animals by tail-cuff plethysmography. Diet: p = 0.9417, Time: p = 0.1382; ND/HSD n = 15 mice/group (Repeated two-way ANOVA and Bonferroni's test). C: HSD does not alter the CBF response to the smooth muscle relaxant adenosine; Diet: p = 0.9659, Time: p = 0.0549; 4 weeks: ND/HSD n = 8/5; 8 weeks: ND/HSD n = 10; 12 weeks: ND/HSD n = 10/8; 24 weeks: ND/HSD n = 7 mice/group (Two-way ANOVA and Tukey's test). D: HSD reduces resting CBF in the hippocampus. * p = 0.0245 vs. 0 weeks; 0 weeks n = 8, 12 weeks n = 7, 24 weeks n = 5 mice/group (One way ANOVA and Tukey’s test). E: Temporal profile of CBF increase induced by whisker stimulation after 4, 8 and 24 weeks of ND or HSD; 4 weeks: ND/HSD n = 7/7; 8 weeks: ND/HSD n = 6/7; 12 weeks: ND/HSD n = 9; 24 weeks: ND/HSD n = 5/7 mice/group. The dotted line indicates the baseline CBF whereas the shaded area represents the standard error. F: CBF increase to whisker stimulation assessed as area under the curve shows a diet effect (Diet: p = 0.0013, Time: p = 0.6687), which did not reach statistical significance at the Tukey’s test; 4 weeks: ND/HSD n = 7/7; 8 weeks: ND/HSD n = 6/7; 12 weeks: ND/HSD n = 9; 24 weeks: ND/HSD n = 5/7 mice/group (Two-way ANOVA and Tukey's test). G: Return to normal diet does not alter MAP and CBF responses to whisker stimulation and adenosine. MAP: p = 0.2406; Whisker: p = 0.2340; Adenosine: p = 0.8010, n = 5 mice/group (One-way ANOVA and Tukey’s test). H: Endothelial dysfunction is also observed in mice fed a 4% HSD for 12 weeks. ACh: * p < 0.0376 vs ND; ND/HSD n = 4/5 mice/group (unpaired t-test, two-tailed). I: A 4% HSD impairs performance at the novel object test (p = 0.0350 vs ND, unpaired t-test, two tailed). J: Resting NO production, assessed by DAF-FM, is reduced in pial microvascular preparation from mice fed a HSD. The effect is reversed by administration of the NO precursor L-arginine. * p = 0.0024; microvessels isolated from 8 (ND Veh), 6 (ND L-Arg), 7 (HSD Veh) and 9 (HSD L-Arg) mice/group (One-way ANOVA and Tukey’s test). Data are expressed as mean ± SEM.

Supplementary Figure 2 Effect of HSD on mRNA levels of proinflammatory mediators in cerebral microvessels.

HSD does not induce upregulation of cytokines, adhesion molecules and other proinflammatory molecules in isolated pial microvascular preparations of mice fed a HSD for 12 weeks. IL-6: * p < 0.0411 vs ND, ND/HSD n = 3/5; VCAM1: * p = 0.0102 vs ND, ND/HSD n = 5 mice/group (unpaired t-test, two-tailed). Data are expressed as mean ± SEM.

Supplementary Figure 3 Effect of HSD or systemic administration of IL-17 on mRNA levels of proinflammatory mediators in brain endothelial cells and on the BBB.

A: HSD or systemic administration of IL-17 do not upregulate cytokines, adhesion molecules and other proinflammatory molecules in endothelial cells FACS-sorted from the brain. ND/HSD n = 10; Veh/IL-17 n = 5/4 mice/group (One-way ANOVA and Tukey’s test). B: HSD does not increase BBB permeability to FITC-dextran (3kDa) both in cortex and hippocampus. p = 0.1143 and p = 0.9624 vs ND; n = 5 mice/group (unpaired t-test, two-tailed). Data are expressed as mean ± SEM.

Supplementary Figure 4 Effect of aging or ETA receptor antagonism on cerebrovascular and cognitive dysfunction induced by HSD.

A: HSD (8 weeks) does not affect MAP in aged mice but, at variance with young mice, significantly attenuates the CBF response induced by whisker stimulation; Diet: * p = 0.0010, Age: * p = 0.0003; ND young/old n = 13-8, HSD young/old n = 12/8 mice/group (Two-way ANOVA and Tukey’s test). HSD does not further decrease the CBF response to ACh in aged mice; Diet: * p < 0.0001, Age: * p = 0.0195 ND young/old n = 10-7, HSD young/old n = 10/8 mice/group (Two-way ANOVA and Tukey’s test). B: Worsening of the performance at the novel object recognition task occurs earlier (8 weeks of HSD), in aged mice than in young mice (12 weeks for HSD)(see Fig. 2a). Total Exploration Time, Diet: * p < 0.0001, Age: p = 0.1935; NOR, Diet: * p = 0.048, Age: * p = 0.0096; ND young/old n = 8–10, HSD young/old n = 10/9 mice/group (Two-way ANOVA and Tukey’s test). C: Neocortical superfusion of the ETA receptor antagonist BQ123 fails to reverse the cerebrovascular effects of HSD. ACh: Diet * p < 0.0008, Treatment p = 0.7523; ND Veh/BQ123 n = 4/4, HSD Veh/BQ123 n = 3/3 mice/group (Two-way ANOVA and Tukey’s test). Data are expressed as mean ± SEM.

Supplementary Figure 5 HSD increases mRNA levels of factors required for Th17 polarization.

A: HSD induces upregulation of IL-22, IL-23R, iNOS, SAA1-3 in distal small intestine of mice fed a HSD. IL-22: * p = 0.0073 vs ND; ND/HSD n = 7/8; IL-23R: * p = 0.0057 vs ND; ND/HSD n = 8/7; iNOS: * p = 0.0005 vs ND; ND/HSD n = 8/9; SAA1: * p = 0.0007 vs ND; ND/HSD n = 8/9; SAA2: * p = 0.0005 vs ND; ND/HSD n = 8/9; SAA3: * p < 0.0001 vs ND; ND/HSD n = 8/9 mice/group (unpaired t-test, two-tailed). B: HSD does not increase IL-17A mRNA levels in the colon. mRNA levels are normalized to IL-17A mRNA levels in the distal small intestine of mice fed a ND. Diet: p = 0.1461, ND/HSD n = 3/4 mice/group (Two-way ANOVA and Tukey's test). C: Plasma levels of TNF-α and IL-6 are not increased after HSD. TNF-α: p = 0.6581 vs ND; IL-6: p = 0.8592 vs ND; n = 12 mice/group (unpaired t-test, two-tailed). n.d: not detectable. Data are expressed as mean ± SEM.

Supplementary Figure 6 HSD does not increase Il17a mRNA levels or TH17 cells in both brain and meninges.

A: HSD does not alter number and B: frequency of T helper and TH17 cells in brain and meninges. Brain T helper: p = 0.1671 vs ND and p = 0.9157 vs ND; Meningeal T helper: p = 0.3882 vs ND and p = 0.5669 vs ND; Brain TH17 cells: p = 0.8709 vs ND and p = 0.6491 vs ND; Meningeal TH17 cells: p = 0.7677 vs ND and p = 0.3192 vs ND; n = 5/group, 2 mice/samples (unpaired t-test, two-tailed). C: IL-17 mRNA levels are not increased in the brain or meninges of mice fed a HSD. Meninges: p = 0.6465 vs ND; ND/HSD n = 4/5 mice group (unpaired t-test, two-tailed). Data are expressed as mean ± SEM.

Supplementary Figure 7 MAP and CBF responses in Il17a–/– or Rag1–/– mice, in WT mice treated with IL-17-neutralizing antibodies and in WT mice treated with IL-17.

A-C: HSD does not alter MAP or CBF responses to whisker stimulation and adenosine in IL17-/- and Rag1-/- mice, or in WT mice receiving IL17 neutralizing antibodies. IL17-/-, MAP, Diet: p = 0.1864, Genotype: p = 0.2153; ND WT/IL17-/- n = 5/4, HSD WT/IL17-/- n = 8/6; Whisker, Diet: p = 0.5651, Genotype: p = 0.8474, WT/IL17-/- n = 6/4, HSD WT/IL17-/- n = 7/6; Adenosine, Diet: p = 0.8710, Genotype: p = 0.1652; WT/IL17-/- n = 6/4, HSD WT/IL17-/- n = 7/7 mice/group. Rag1-/-, MAP, Diet: p = 0.0619, Genotype: p = 0.4521; ND WT/Rag1-/- n = 4/8, HSD WT/Rag1-/- n = 8/10; Whisker, Diet: * p = 0.0094, Genotype: p = 0.5461, WT/Rag1-/- n = 5/8, HSD WT/Rag1-/- n = 8/8; Adenosine, Diet: p = 0.6543, Genotype: p = 0.7647; WT/Rag1-/- n = 5/7, HSD WT/Rag1-/- n = 8/9 mice/group (Two-way ANOVA and Tukey’s test). D: Administration of IL-17 in WT mice does not affect MAP or the increase in CBF produced by adenosine; MAP: p = 0.7772 vs Veh; Veh/IL-17 n = 7-8/mice; Adenosine: p = 0.7102 vs Veh; Veh/IL-17 n = 7-8/mice group (unpaired t-test, two-tailed). Data are expressed as mean ± SEM.

Supplementary Figure 8 Effect of depletion of brain perivascular macrophages and of FTY720 or Y27632 on CBF responses in mice fed HSD.

A: PVM depletion by clodronate does not ameliorate the endothelial dysfunction in mice fed a HSD diet; Diet: * p < 0.0001, Treatment: p = 0.8889; ND Veh/ND CLO n = 4, HSD Veh/HSD CLO n = 5 mice/group (Two-way ANOVA and Tukey’s test). B: Clodronate (i.c.v.) depletes brain PVM in the somatosensory cortex of both ND and HSD-fed mice; Diet: p = 0.9780, Treatment: * p < 0.0001; ND Veh/ND CLO n = 4, HSD Veh/HSD CLO n = 5 mice/group (Two-way ANOVA and Tukey’s test). C: FTY720 has no effect on the endothelial dysfunction induced by HSD; Diet: * p = 0.0001, Treatment: p = 0.3789; ND Veh/ND FTY720 n = 6/4, HSD Veh/HSD FTY720 n = 6/5 mice/group (Two-way ANOVA and Tukey’s test). D: FTY720 administration reduces blood T-helper lymphocytes in both ND and HSD-fed mice. Diet: p = 0.7357, Treatment: p < 0.0001; ND Veh/ND FTY720 n = 6/3, HSD Veh/HSD FTY720 n = 5/4 mice/group (Two-way ANOVA and Tukey’s test). E: The CBF responses to whisker stimulation or adenosine are not altered by Y27632; p > 0.05 vs ND; Whisker, Diet: p = 0.5397, Treatment: p = 0.5804; ND Veh/Y27632 n = 6/6, HSD Veh/Y27632 n = 6/8; Adenosine, Diet: p = 0.5712, Treatment: p = 0.4964; ND Veh/Y27632 n = 6/6, HSD Veh/Y27632 n = 6/7 mice/group (Two-way ANOVA and Tukey’s test). Data are expressed as mean ± SEM.

Supplementary Figure 9 Effect of IL-17 administration on eNOS phosphorylation in human cerebral endothelial cells (HBEC.5i).

A: IL-17 (1-10 ng/mL) increases phosphorylation of eNOS on Thr495, an effect abrogated by the administration of the ROCK inhibitor Y27632 (5μM). * p = 0.0026; Vehicle: IL-17 0 ng/ml n = 8; IL-17 1 ng/ml n = 7, IL-17 10 ng/ml n = 5; Y27632: IL-17 0-10 ng/ml n = 4 independent experiments/group (One-way ANOVA and Tukey’s test). B-D: HSD increases the inhibitory phosphorylation of eNOS at Thr495 in isolated pial microvascular preparations isolated from WT mice and mice injected with an IgG control antibody or vehicle. WT: * p = 0.0380 vs ND; ND/HSD n = 3/6; IgG: * p = 0.0272 vs ND; ND/HSD n = 5/6; Veh: * p = 0.0177 vs ND; ND/HSD n = 3/4 mice/group (unpaired t-test, two-tailed). Data are expressed as mean ± SEM. Immunoblots in A, B, C and D are cropped. Full gel pictures for immunoblots are shown in Supplementary Fig 14.

Supplementary Figure 10 Effect of IL-17 and TNF-α administration on mRNA levels of proinflammatory mediators in brain endothelial cells (bEnd.3).

A: IL-17 (10 ng/mL) does not induce upregulation of cytokines, adhesion molecules and other pro-inflammatory molecules in cultures of bEnd.3 cells. p > 0.05 vs 0; n = 4 independent experiments/group (One-way ANOVA and Tukey’s test). B: TNF-α (10 ng/mL) induces upregulation of cytokines and adhesion molecules in cultures of bEnd.3 cells. ICAM1: * p = 0.0124 vs 0; VCAM1: * p < 0.0001 vs 0; CXCL1: * p = 0.0024; CXCL2: * p = 0.0035 vs 0; CXCL5: * p = 0.0372 vs 0; MCP1: * p < 0.0001 vs 0; n = 3 independent experiments/group (One-way ANOVA and Tukey’s test). Data are expressed as mean ± SEM.

Supplementary Figure 11 Cartoon illustrating the mechanisms of neurovascular and cognitive dysfunction induced by HSD.

HSD induces a TH17 response in the distal small intestine by presumably activating serum glucocorticoid-regulated kinase 1 (SGK1). TH17 cells lead to an increase in circulating IL17, which, in turn, acts on cerebral endothelial cells to induce ROCK activation, inhibitory eNOS phosphorylation, and reduction in eNOS catalytic activity. The resulting reduction in endothelial NO leads to cerebral hypoperfusion, vascular dysregulation and cognitive impairment.

Supplementary Figure 12 Full immunoblots image of panels in Figs. 3 and 6.

Membranes were incubated with anti-pThr495 eNOS antibody (1:500) or anti-pSer1177 eNOS antibody (1:1000) overnight. After incubation with secondary antibody, protein bands were visualized with Super Signal West Femto Maximum Sensitivity Substrate (Thermo Fisher Scientific) on a Bio Rad ChemiDoc MP Imaging System. After washes with TBS/0.1% Tween-20 (TBST), membranes were incubated with anti-eNOS antibody (1:1000) overnight. eNOS bands were visualized with Clarity Western ECL Substrate (BioRad). Samples from microvessels preparations (≈30 µg of protein/lane).

Supplementary Figure 13 Full immunoblots image of panels in Figs. 7 and 8.

Membranes were incubated with anti-pThr495 eNOS antibody (1:500) overnight. After incubation with secondary antibody, protein bands were visualized with Super Signal West Femto Maximum Sensitivity Substrate (Thermo Fisher Scientific) on a Bio Rad ChemiDoc MP Imaging System. After washes with TBS/0.1% Tween-20 (TBST), membranes were incubated with anti-eNOS antibody (1:1000) overnight. eNOS bands were visualized with Clarity Western ECL Substrate (BioRad). Samples from microvessels preparations and cultures of mouse brain endothelial cells (≈30µg of protein/lane).

Supplementary Figure 14 Full immunoblots image of panels in Supplementary Figure 9.

Membranes were incubated with anti-pThr495 eNOS antibody (1:500) overnight. After incubation with secondary antibody, protein bands were visualized with Super Signal West Femto Maximum Sensitivity Substrate (Thermo Fisher Scientific) on a Bio Rad ChemiDoc MP Imaging System. After washes with TBS/0.1% Tween-20 (TBST), membranes were incubated with anti-eNOS antibody (1:1000) overnight. eNOS bands were visualized with Clarity Western ECL Substrate (BioRad). Samples from microvessels preparations and cultures of human brain endothelial cells (≈30 µg of protein/lane).

Supplementary Figure 15 Flow cytometry gating strategy for IL-17+ cells.

Cells were discriminated by FSC-SSC gating and then gated for CD45+CD4+TCRγδ-IL17+ (Th17 cells) or CD45+CD4-TCRγδ+IL17+ (IL17+γδ T cells).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Tables 1 and 2

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Faraco, G., Brea, D., Garcia-Bonilla, L. et al. Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response. Nat Neurosci 21, 240–249 (2018). https://doi.org/10.1038/s41593-017-0059-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-017-0059-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