Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Brent Shell is active.

Publication


Featured researches published by Brent Shell.


Neuron | 2015

High Salt Intake Increases Blood Pressure via BDNF-Mediated Downregulation of KCC2 and Impaired Baroreflex Inhibition of Vasopressin Neurons

Katrina Y. Choe; Su Y. Han; Perrine Gaub; Brent Shell; Daniel L. Voisin; Blayne A. Knapp; Philip A. Barker; Colin H. Brown; J. Thomas Cunningham; Charles W. Bourque

The mechanisms by which dietary salt promotes hypertension are unknown. Previous work established that plasma [Na(+)] and osmolality rise in proportion with salt intake and thus promote release of vasopressin (VP) from the neurohypophysis. Although high levels of circulating VP can increase blood pressure, this effect is normally prevented by a potent GABAergic inhibition of VP neurons by aortic baroreceptors. Here we show that chronic high salt intake impairs baroreceptor inhibition of rat VP neurons through a brain-derived neurotrophic factor (BDNF)-dependent activation of TrkB receptors and downregulation of KCC2 expression, which prevents inhibitory GABAergic signaling. We show that high salt intake increases the spontaneous firing rate of VP neurons in vivo and that circulating VP contributes significantly to the elevation of arterial pressure under these conditions. These results provide the first demonstration that dietary salt can affect blood pressure through neurotrophin-induced plasticity in a central homeostatic circuit.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2013

Central losartan attenuates increases in arterial pressure and expression of FosB/ΔFosB along the autonomic axis associated with chronic intermittent hypoxia

W. David Knight; Ashwini Saxena; Brent Shell; T. Prashant Nedungadi; Steven W. Mifflin; J. Thomas Cunningham

Chronic intermittent hypoxia (CIH) increases mean arterial pressure (MAP) and FosB/ΔFosB staining in central autonomic nuclei. To test the role of the brain renin-angiotensin system (RAS) in CIH hypertension, rats were implanted with intracerebroventricular (icv) cannulae delivering losartan (1 μg/h) or vehicle (VEH) via miniosmotic pumps and telemetry devices for arterial pressure recording. A third group was given the same dose of losartan subcutaneously (sc). Two groups of losartan-treated rats served as normoxic controls. Rats were exposed to CIH or normoxia for 7 days and then euthanized for immunohistochemistry. Intracerebroventricular losartan attenuated CIH-induced increases in arterial pressure during CIH exposure (0800-1600 during the light phase) on days 1, 6, and 7 and each day during the normoxic dark phase. FosB/ΔFosB staining in the organum vasculosum of the lamina terminalis (OVLT), median preoptic nucleus (MnPO), paraventricular nucleus of the hypothalamus (PVN), the rostral ventrolateral medulla (RVLM), and the nucleus of the solitary tract (NTS) was decreased in icv losartan-treated rats. Subcutaneous losartan also reduced CIH hypertension during the last 2 days of CIH and produced bradycardia prior to the effect on blood pressure. Following sc losartan, FosB/ΔFosB staining was reduced only in the OVLT, MnPO, PVN, and NTS. These data indicate that the central and peripheral RAS contribute to CIH-induced hypertension and transcriptional activation of autonomic nuclei and that the contribution of the central RAS is greater during the normoxic dark phase of CIH hypertension.


Current Hypertension Reports | 2016

Neural Control of Blood Pressure in Chronic Intermittent Hypoxia.

Brent Shell; Katelynn Faulk; J. Thomas Cunningham

Sleep apnea (SA) is increasing in prevalence and is commonly comorbid with hypertension. Chronic intermittent hypoxia is used to model the arterial hypoxemia seen in SA, and through this paradigm, the mechanisms that underlie SA-induced hypertension are becoming clear. Cyclic hypoxic exposure during sleep chronically stimulates the carotid chemoreflexes, inducing sensory long-term facilitation, and drives sympathetic outflow from the hindbrain. The elevated sympathetic tone drives hypertension and renal sympathetic activity to the kidneys resulting in increased plasma renin activity and eventually angiotensin II (Ang II) peripherally. Upon waking, when respiration is normalized, the sympathetic activity does not diminish. This is partially because of adaptations leading to overactivation of the hindbrain regions controlling sympathetic outflow such as the nucleus tractus solitarius (NTS), and rostral ventrolateral medulla (RVLM). The sustained sympathetic activity is also due to enhanced synaptic signaling from the forebrain through the paraventricular nucleus (PVN). During the waking hours, when the chemoreceptors are not exposed to hypoxia, the forebrain circumventricular organs (CVOs) are stimulated by peripherally circulating Ang II from the elevated plasma renin activity. The CVOs and median preoptic nucleus chronically activate the PVN due to the Ang II signaling. All together, this leads to elevated nocturnal mean arterial pressure (MAP) as a response to hypoxemia, as well as inappropriately elevated diurnal MAP in response to maladaptations.


Physiological Reports | 2017

Chronic intermittent hypoxia induces oxidative stress and inflammation in brain regions associated with early‐stage neurodegeneration

Brina Snyder; Brent Shell; J. Thomas Cunningham; Rebecca L. Cunningham

Sleep apnea is a common comorbidity of neurodegenerative diseases, such as Alzheimers disease (AD) and Parkinsons disease (PD). Previous studies have shown an association between elevated oxidative stress and inflammation with severe sleep apnea. Elevated oxidative stress and inflammation are also hallmarks of neurodegenerative diseases. We show increased oxidative stress and inflammation in a manner consistent with early stages of neurodegenerative disease in an animal model of mild sleep apnea. Male rats were exposed to 7 days chronic intermittent hypoxia (CIH) for 8 h/day during the light period. Following CIH, plasma was collected and tested for circulating oxidative stress and inflammatory markers associated with proinflammatory M1 or anti‐inflammatory M2 profiles. Tissue punches from brain regions associated with different stages of neurodegenerative diseases (early stage: substantia nigra and entorhinal cortex; intermediate: hippocampus; late stage: rostral ventrolateral medulla and solitary tract nucleus) were also assayed for inflammatory markers. A subset of the samples was examined for 8‐hydroxydeoxyguanosine (8‐OHdG) expression, a marker of oxidative stress‐induced DNA damage. Our results showed increased circulating oxidative stress and inflammation. Furthermore, brain regions associated with early‐stage (but not late‐stage) AD and PD expressed oxidative stress and inflammatory profiles consistent with reported observations in preclinical neurodegenerative disease populations. These results suggest mild CIH induces key features that are characteristic of early‐stage neurodegenerative diseases and may be an effective model to investigate mechanisms contributing to oxidative stress and inflammation in those brain regions.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2017

Role of angiotensin-converting enzyme 1 within the median preoptic nucleus following chronic intermittent hypoxia

Katelynn Faulk; Brent Shell; T. Prashant Nedungadi; J. Thomas Cunningham

Sustained hypertension is an important consequence of obstructive sleep apnea. An animal model of the hypoxemia associated with sleep apnea, chronic intermittent hypoxia (CIH), produces increased sympathetic nerve activity (SNA) and sustained increases in blood pressure. Many mechanisms have been implicated in the hypertension associated with CIH, including the role of ΔFosB within the median preoptic nucleus (MnPO). Also, the renin-angiotensin system (RAS) has been associated with CIH hypertension. We conducted experiments to determine the possible association of FosB/ΔFosB with a RAS component, angiotensin-converting enzyme 1 (ACE1), within the MnPO following 7 days of CIH. Retrograde tract tracing from the paraventricular nucleus (PVN), a downstream region of the MnPO, was used to establish a potential pathway for FosB/ΔFosB activation of MnPO ACE1 neurons. After CIH, ACE1 cells with FosB/ΔFosB expression increased colocalization with a retrograde tracer that was injected unilaterally within the PVN. Also, Western blot examination showed ACE1 protein expression increasing within the MnPO following CIH. Chromatin immunoprecipitation (ChIP) assays demonstrated an increase in FosB/ΔFosB association with the ACE1 gene within the MnPO following CIH. FosB/ΔFosB may transcriptionally target ACE1 within the MnPO following CIH to affect the downstream PVN region, which may influence SNA and blood pressure.


Archive | 2016

The Role of Angiotensin II in Central Control of Blood Pressure and Body Fluid Homeostasis

Brent Shell


The FASEB Journal | 2015

AT1a Receptor Upregulation in the MnPO Following CIH Leads to Sustained Hypertension

Brent Shell; Thekkethil Prashant Nedungadi; J. Cunningham


Archive | 2015

Median Preoptic AT1a Receptor Increase Responsible for Sustained Component of Hypertension from Chronic Intermittent Hypoxia

Brent Shell; T. Prashant Nedungadi; J. Thomas Cunningham


Archive | 2015

Chronic Intermittent Hypoxia increases oxidative stress and inflammation

Brina Snyder; Brent Shell; J. Thomas Cunningham; Rebecca L. Cunningham


Archive | 2015

ratssystemic hypertension in conscious baroreflex sensitivity are associated with intermittent Enhanced sympathetic outflow and decreased

C. J. Lai; Christopher Yang; Y. Y. Hsu; Y. N. Lin; T. B. J. Kuo; Amanda L. Sharpe; Mary Ann Andrade; Myrna Herrera-Rosales; Steven L. Britton; G Lauren; J. Thomas Cunningham; W. David Knight; Ashwini Saxena; Brent Shell; T. Prashant Nedungadi; Steven W. Mifflin

Collaboration


Dive into the Brent Shell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

T. Prashant Nedungadi

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

J. Cunningham

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Ashwini Saxena

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Brina Snyder

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Katelynn Faulk

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Rebecca L. Cunningham

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Steven W. Mifflin

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

W. David Knight

University of North Texas

View shared research outputs
Top Co-Authors

Avatar

Charles W. Bourque

McGill University Health Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge