Salt consumption worldwide greatly exceeds minimal requirements, and excessive dietary salt has emerged as a powerful risk factor for cardiovascular diseases, especially stroke. A high salt diet (HSD) is particularly damaging in older people. Although the deleterious effect of HSD was first attributed to the hypertension that develops in salt-sensitive individuals, increasing evidence indicates that dietary salt increases the incidence of stroke independently of whether or not the blood pressure is elevated. However, it remains to be established how HSD acts on the brain to increase cerebrovascular risk independently of hypertension. International attempts to curb salt consumption have met with limited success and there is an ongoing controversy about ideal levels of salt intake. Therefore, there have been calls for gaining a better mechanistic understanding of how dietary salt impacts the vascular health of the brain. Recent evidence indicates that HSD induces the expansion of a select population of T-helper lymphocytes in the gut (Th17 cells), which produce IL17, a cytokine well known for its damaging vascular effects in animals and humans. Indeed, diseases driven by a Th17 immune response, such as rheumatoid arthritis, psoriasis, multiple sclerosis or inflammatory bowel disease, are associated with increased risk of stroke. On these bases, we propose to test the central hypothesis that HSD exerts its deleterious cerebrovascular effects by inducing a Th17 response. The resulting increase in the vasotoxic cytokine IL17, in turn, acts on cerebral blood vessels to alter critical homeostatic responses that control cerebral perfusion and safeguard brain health. To this end, the present grant application will examine neurovascular function in a mouse model of HSD (4 or 8% NaCl for 8 weeks) to test the following hypotheses in young and aging mice: (1) HSD alters microvascular structure and function to disrupt key cerebrovascular regulatory mechanisms that assure that the brain receives sufficient blood flow well matched to its energetic needs; (2) The cerebrovascular dysfunction is mediated by Th17 lymphocytes via the cytokine IL17; (3) IL17, in turn, causes cerebrovascular dysfunction by inducing NADPH oxidase-dependent oxidative stress, as well as by modulating the phosphorylation state of endothelial nitric oxide synthase and suppressing nitric oxide production. The proposed studies fill an obvious gap in the understanding of the cerebrovascular effects of HSD across the lifespan, and open new avenues of translational research to develop rational strategies to contain the impact of salt on brain vascular health. Furthermore, the concept that Th17 responses initiated in other organs may alter cerebrovascular structure and function has far reaching consequences for Th17-dependent diseases associated with increased stroke risk.

Public Health Relevance

Stroke remains a leading cause of mortality and morbidity worldwide and high salt consumption has emerged as a major contributor to the risk of developing stroke. The present application seeks to explore the novel hypothesis that the deleterious effects of a diet rich in salt are mediated by an immune response induced by the salt, which, in turn, damages cerebral blood vessels. The proposed studies open the way to future research to develop approaches to contain the harmful effects of salt on brain vascular health.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS095441-04
Application #
9593519
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Koenig, James I
Project Start
2015-12-01
Project End
2020-11-30
Budget Start
2018-12-01
Budget End
2019-11-30
Support Year
4
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Weill Medical College of Cornell University
Department
Neurology
Type
Schools of Medicine
DUNS #
060217502
City
New York
State
NY
Country
United States
Zip Code
10065
Faraco, Giuseppe; Brea, David; Garcia-Bonilla, Lidia et al. (2018) Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response. Nat Neurosci 21:240-249
Iadecola, Costantino; Gottesman, Rebecca F (2018) Cerebrovascular Alterations in Alzheimer Disease. Circ Res 123:406-408
Mallat, Ziad; Ait-Oufella, Hafid; Tedgui, Alain (2017) The Pathogenic Transforming Growth Factor-? Overdrive Hypothesis in Aortic Aneurysms and Dissections: A Mirage? Circ Res 120:1718-1720
Gorelick, Philip B; Furie, Karen L; Iadecola, Costantino et al. (2017) Defining Optimal Brain Health in Adults: A Presidential Advisory From the American Heart Association/American Stroke Association. Stroke 48:e284-e303
Merkler, Alexander E; Iadecola, Costantino (2017) Rollercoaster Blood Pressure: An Alzheimer Disease Risk Factor? Circulation 136:526-528
Iadecola, Costantino (2017) The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron 96:17-42
Gusdon, Aaron M; Gialdini, Gino; Kone, Gbambele et al. (2017) Neutrophil-Lymphocyte Ratio and Perihematomal Edema Growth in Intracerebral Hemorrhage. Stroke 48:2589-2592
Gialdini, Gino; Parikh, Neal S; Chatterjee, Abhinaba et al. (2017) Rates of Spinal Cord Infarction After Repair of Aortic Aneurysm or Dissection. Stroke 48:2073-2077
Park, Laibaik; Uekawa, Ken; Garcia-Bonilla, Lidia et al. (2017) Brain Perivascular Macrophages Initiate the Neurovascular Dysfunction of Alzheimer A? Peptides. Circ Res 121:258-269
Yanagida, Keisuke; Liu, Catherine H; Faraco, Giuseppe et al. (2017) Size-selective opening of the blood-brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci U S A 114:4531-4536

Showing the most recent 10 out of 14 publications