Sleep apnea (SA) affects ~15% of the US population. Most people with SA develop neurogenic hypertension (HTN) associated with elevated sympathetic nerve activity (SNA). During the previous funding cycle, we modeled SA HTN by exposing rats to chronic intermittent hypoxia (CIH). We determined that HTN induced by 7 days of CIH is maintained by pre-sympathetic PVN neuronal discharge driven by exaggerated NMDA receptor (NMDAR) tone. Importantly, we discovered that PVN activation by CIH involves local adaptive responses (i.e., plasticity) wherein expression of neuronal NMDAR subunits (GluN1 & GluN2B) is reduced and expression of the glial L-glutamate (L-Glu) transporter EEAT2 is increased. These responses reflect homeostatic adaptations to exaggerated glutamatergic input, which we postulate arises, at least in part, from the hindbrain NTS (Project 1) and the forebrain MnPO (Project 2). CIH also decreased PVN expression of the adaptor protein PSD95 that forms a ternary complex with the NMDAR GluN2B subunit and neuronal nitric oxide (NO) synthase (nNOS). This complex couples NMDAR Ca2+ influx with production of NO. We hypothesize that these adaptations to CIH have two offsetting actions. (1) Reduced NMDAR and increased EAAT2 expression blunt glutamatergic PVN activation and thereby buffer development of HTN. (2) Reduced PSD95 blunts NMDAR-driven NO production and lessens its tonic facilitation of GABA release. This disinhibits the PVN and thereby supports development of HTN. The net effect of these opposing adaptations is that HTN induced by CIH is less pronounced than it would be in their absence. In addition to HTN, SA increases the risk of ischemic stroke by ~4 fold. The same PVN adaptations to SA/CIH that participate in development of HTN are hypothesized to reduce ischemic injury by limiting the rise of extracellular L-Glu and reducing the production of neurotoxic NO. Studies in this revised proposal will expose rats to 7 and 28 days of CIH and CIH with hypercapnia (CIHHC) to more closely model human SA.
Our specific aims will: (1) Determine effects of CIH & CIHHC on neurogenic HTN, PVN expression of synaptic/excitotoxic signaling proteins and PVN neuronal/tissue survival after local ischemia. (2) Determine effects of CIH/CIHHC adaptations on PVN control of SNA/MAP and mechanisms of neuronal vulnerability to local ischemia. (3) Use viral-mediated gene transfer/shRNA knockdown to mimic and rescue specific PVN adaptations and determine their contributions to hypertensive and neuroprotective effects of CIH & CIHHC.

Public Health Relevance

Sleep apnea (SA) is commonly linked to systemic hypertension. It also increases the risk of stroke by ~4 fold. By exposing rats to intermittent hypoxia with and without concurrent hypercapnia, we will model SA and map the resulting neuroadaptations. In doing so, we will determine how individual neuroadaptations contribute both to SA-associated hypertension and to neuronal vulnerability/protection from ischemia.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program Projects (P01)
Project #
5P01HL088052-07
Application #
9096159
Study Section
Special Emphasis Panel (ZHL1)
Project Start
Project End
Budget Start
2016-04-01
Budget End
2017-03-31
Support Year
7
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of North Texas
Department
Type
DUNS #
110091808
City
Fort Worth
State
TX
Country
United States
Zip Code
76107
Mitchell, N C; Gilman, T L; Daws, L C et al. (2018) High salt intake enhances swim stress-induced PVN vasopressin cell activation and active stress coping. Psychoneuroendocrinology 93:29-38
Wu, Qiong; Cunningham, J Thomas; Mifflin, Steve (2018) Transcription factor ?FosB acts within the nucleus of the solitary tract to increase mean arterial pressure during exposures to intermittent hypoxia. Am J Physiol Heart Circ Physiol 314:H270-H277
Holbein, Walter W; Blackburn, Megan B; Andrade, Mary Ann et al. (2018) Burst patterning of hypothalamic paraventricular nucleus-driven sympathetic nerve activity in ANG II-salt hypertension. Am J Physiol Heart Circ Physiol 314:H530-H541
Blackburn, Megan B; Andrade, Mary Ann; Toney, Glenn M (2018) Hypothalamic PVN contributes to acute intermittent hypoxia-induced sympathetic but not phrenic long-term facilitation. J Appl Physiol (1985) 124:1233-1243
Wang, Lei A; Nguyen, Dianna H; Mifflin, Steve W (2018) CRHR2 (Corticotropin-Releasing Hormone Receptor 2) in the Nucleus of the Solitary Tract Contributes to Intermittent Hypoxia-Induced Hypertension. Hypertension 72:994-1001
Lalley, Peter M; Mifflin, Steve W (2017) Oscillation patterns are enhanced and firing threshold is lowered in medullary respiratory neuron discharges by threshold doses of a ?-opioid receptor agonist. Am J Physiol Regul Integr Comp Physiol 312:R727-R738
Snyder, Brina; Shell, Brent; Cunningham, J Thomas et al. (2017) Chronic intermittent hypoxia induces oxidative stress and inflammation in brain regions associated with early-stage neurodegeneration. Physiol Rep 5:
Granato, Álisson Silva; Gomes, Paula Magalhães; Martins Sá, Renato William et al. (2017) Cardiovascular responses to l-glutamate microinjection into the NTS are abrogated by reduced glutathione. Neurosci Lett 642:142-147
Faulk, Katelynn E; Nedungadi, T Prashant; Cunningham, J Thomas (2017) Angiotensin converting enzyme 1 in the median preoptic nucleus contributes to chronic intermittent hypoxia hypertension. Physiol Rep 5:
Faulk, Katelynn; Shell, Brent; Nedungadi, T Prashant et al. (2017) Role of angiotensin-converting enzyme 1 within the median preoptic nucleus following chronic intermittent hypoxia. Am J Physiol Regul Integr Comp Physiol 312:R245-R252

Showing the most recent 10 out of 37 publications