Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 317(5): H1002-H1012, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31469293

ABSTRACT

Percutaneous creation of a small central arteriovenous (AV) fistula is currently being evaluated for the treatment of uncontrolled hypertension (HT). Although the mechanisms that contribute to the antihypertensive effects of the fistula are unclear, investigators have speculated that chronic blood pressure (BP) lowering may be due to 1) reduced total peripheral resistance (TPR), 2) increased secretion of atrial natriuretic peptide (ANP), and/or 3) suppression of renal sympathetic nerve activity (RSNA). We used an established integrative mathematical model of human physiology to investigate these possibilities from baseline conditions that mimic sympathetic overactivity and impaired renal function in patients with resistant HT. After a small fistula was stimulated, there were sustained increases in cardiac output, atrial pressures, and plasma ANP concentration (3-fold), without suppression of RSNA; at 8 wk, BP was reduced 14 mmHg along with a 32% fall in TPR. In contrast, when this simulation was repeated while clamping ANP at baseline BP decreased only 4 mmHg, despite a comparable fall in TPR. Furthermore, when chronic resetting of atrial mechanoreceptors was prevented during the fistula, RSNA decreased 7%, and along with the same threefold increase in ANP, BP fell 19 mmHg. This exaggerated fall in BP occurred with a similar decrease in TPR when compared with the above simulations. These findings suggest that ANP, but not TPR, is a key determinant of long-term BP lowering after the creation of an AV fistula and support a contribution of suppressed RSNA if resetting of the atrial-renal reflex is truly incomplete.NEW & NOTEWORTHY The mechanisms that contribute to the antihypertensive effects of a small arteriovenous (AV) fistula comparable to the size used by the ROX coupler currently in clinical trials are unclear and not readily testable in clinical or experimental studies. The integrative mathematical model of human physiology used in the current study provides a tool for understanding key causal relationships that account for blood pressure (BP) lowering and for testing competing hypotheses. The findings from the simulations suggest that after creation of a small AV fistula increased ANP secretion plays a critical role in mediating long-term reductions in BP. Measurement of natriuretic peptide levels in hypertensive patients implanted with the ROX coupler would provide one critical test of this hypothesis.


Subject(s)
Arteriovenous Shunt, Surgical , Atrial Pressure , Blood Pressure , Cardiac Output , Heart Atria/innervation , Hypertension/surgery , Kidney/innervation , Mechanoreceptors/metabolism , Models, Cardiovascular , Sympathetic Nervous System/physiopathology , Antihypertensive Agents/therapeutic use , Atrial Natriuretic Factor/blood , Blood Pressure/drug effects , Computer Simulation , Drug Resistance , Humans , Hypertension/blood , Hypertension/physiopathology , Reflex , Time Factors
2.
J Am Coll Cardiol ; 73(23): 3006-3017, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31196459

ABSTRACT

Three recent renal denervation studies in both drug-naïve and drug-treated hypertensive patients demonstrated a significant reduction of ambulatory blood pressure compared with respective sham control groups. Improved trial design, selection of relevant patient cohorts, and optimized interventional procedures have likely contributed to these positive findings. However, substantial variability in the blood pressure response to renal denervation can still be observed and remains a challenging and important problem. The International Sympathetic Nervous System Summit was convened to bring together experts in both experimental and clinical medicine to discuss the current evidence base, novel developments in our understanding of neural interplay, procedural aspects, monitoring of technical success, and others. Identification of relevant trends in the field and initiation of tailored and combined experimental and clinical research efforts will help to address remaining questions and provide much-needed evidence to guide clinical use of renal denervation for hypertension treatment and other potential indications.


Subject(s)
Blood Pressure Monitoring, Ambulatory/trends , Congresses as Topic/trends , Hypertension/surgery , Internationality , Kidney/innervation , Sympathectomy/trends , Blood Pressure/physiology , Denervation/methods , Denervation/trends , Humans , Hypertension/diagnosis , Hypertension/physiopathology , Kidney/physiology , Randomized Controlled Trials as Topic/methods , Review Literature as Topic , Sympathectomy/methods , Sympathetic Nervous System/physiology , Sympathetic Nervous System/physiopathology
3.
Circ Res ; 124(7): 1071-1093, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30920919

ABSTRACT

Despite availability of effective drugs for hypertension therapy, significant numbers of hypertensive patients fail to achieve recommended blood pressure levels on ≥3 antihypertensive drugs of different classes. These individuals have a high prevalence of adverse cardiovascular events and are defined as having resistant hypertension (RHT) although nonadherence to prescribed antihypertensive medications is common in patients with apparent RHT. Furthermore, apparent and true RHT often display increased sympathetic activity. Based on these findings, technology was developed to treat RHT by suppressing sympathetic activity with electrical stimulation of the carotid baroreflex and catheter-based renal denervation (RDN). Over the last 15 years, experimental and clinical studies have provided better understanding of the physiological mechanisms that account for blood pressure lowering with baroreflex activation and RDN and, in so doing, have provided insight into which patients in this heterogeneous hypertensive population are most likely to respond favorably to these device-based therapies. Experimental studies have also played a role in modifying device technology after early clinical trials failed to meet key endpoints for safety and efficacy. At the same time, these studies have exposed potential differences between baroreflex activation and RDN and common challenges that will likely impact antihypertensive treatment and clinical outcomes in patients with RHT. In this review, we emphasize physiological studies that provide mechanistic insights into blood pressure lowering with baroreflex activation and RDN in the context of progression of clinical studies, which are now at a critical point in determining their fate in RHT management.


Subject(s)
Baroreflex , Blood Pressure , Catheter Ablation/instrumentation , Drug Resistance , Electric Stimulation Therapy/instrumentation , Hypertension/therapy , Implantable Neurostimulators , Sympathectomy/instrumentation , Sympathetic Nervous System/physiopathology , Animals , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Catheter Ablation/adverse effects , Electric Stimulation Therapy/adverse effects , Equipment Design , Humans , Hypertension/diagnosis , Hypertension/epidemiology , Hypertension/physiopathology , Sympathectomy/adverse effects , Treatment Outcome
4.
Am J Physiol Heart Circ Physiol ; 315(5): H1368-H1382, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30004810

ABSTRACT

Electrical stimulation of the baroreflex chronically suppresses sympathetic activity and arterial pressure and is currently being evaluated for the treatment of resistant hypertension. The antihypertensive effects of baroreflex activation are often attributed to renal sympathoinhibition. However, baroreflex activation also decreases heart rate, and robust blood pressure lowering occurs even after renal denervation. Because controlling renal sympathetic nerve activity (RSNA) and cardiac autonomic activity cannot be achieved experimentally, we used an established mathematical model of human physiology (HumMod) to provide mechanistic insights into their relative and combined contributions to the cardiovascular responses during baroreflex activation. Three-week responses to baroreflex activation closely mimicked experimental observations in dogs including decreases in blood pressure, heart rate, and plasma norepinephrine and increases in plasma atrial natriuretic peptide (ANP), providing validation of the model. Simulations showed that baroreflex-induced alterations in cardiac sympathetic and parasympathetic activity lead to a sustained depression of cardiac function and increased secretion of ANP. Increased ANP and suppression of RSNA both enhanced renal excretory function and accounted for most of the chronic blood pressure lowering during baroreflex activation. However, when suppression of RSNA was blocked, the blood pressure response to baroreflex activation was not appreciably impaired due to inordinate fluid accumulation and further increases in atrial pressure and ANP secretion. These simulations provide a mechanistic understanding of experimental and clinical observations showing that baroreflex activation effectively lowers blood pressure in subjects with previous renal denervation. NEW & NOTEWORTHY Both experimental and clinical studies have shown that the presence of renal nerves is not an obligate requirement for sustained reductions in blood pressure during chronic electrical stimulation of the carotid baroreflex. Simulations using HumMod, a mathematical model of integrative human physiology, indicated that both increased secretion of atrial natriuretic peptide and suppressed renal sympathetic nerve activity play key roles in mediating long-term reductions in blood pressure during chronic baroreflex activation.


Subject(s)
Arterial Pressure , Autonomic Nervous System/physiopathology , Baroreflex , Computer Simulation , Heart Rate , Heart/innervation , Hypertension/physiopathology , Kidney/innervation , Models, Cardiovascular , Pressoreceptors/physiopathology , Animals , Atrial Natriuretic Factor/blood , Autonomic Nervous System/metabolism , Dogs , Electric Stimulation Therapy , Humans , Hypertension/blood , Hypertension/therapy , Models, Animal , Norepinephrine/blood , Sympathectomy , Time Factors
5.
Front Physiol ; 9: 455, 2018.
Article in English | MEDLINE | ID: mdl-29760664

ABSTRACT

Aim: Activation of the sympathetic nervous system is common in resistant hypertension (RHT) and also in chronic kidney disease (CKD), a prevalent condition among resistant hypertensives. However, renal nerve ablation lowers blood pressure (BP) only in some patients with RHT. The influence of loss of nephrons per se on the antihypertensive response to renal denervation (RDNx) is unclear and was the focus of this study. Methods: Systemic hemodynamics and sympathetically mediated low frequency oscillations of systolic BP were determined continuously from telemetrically acquired BP recordings in rats before and after surgical excision of ∼80% of renal mass and subsequent RDNx. Results: After reduction of renal mass, rats fed a high salt (HS) diet showed sustained increases in mean arterial pressure (108 ± 3 mmHg to 128 ± 2 mmHg) and suppression of estimated sympathetic activity (∼15%), responses that did not occur with HS before renal ablation. After denervation of the remnant kidney, arterial pressure fell (to 104 ± 4 mmHg), estimated sympathetic activity and heart rate (HR) increased concomitantly, but these changes gradually returned to pre-denervation levels over 2 weeks of follow up. Subsequently, sympathoinhibition with clonidine did not alter arterial pressure while significantly suppressing estimated sympathetic activity and HR. Conclusion: These results indicate that RDNx does not chronically lower arterial pressure in this model of salt-sensitive hypertension associated with substantial nephron loss, but without ischemia and increased sympathetic activity, thus providing further insight into conditions likely to impact the antihypertensive response to renal-specific sympathoinhibition in subjects with CKD.

6.
Hypertension ; 68(6): 1400-1406, 2016 12.
Article in English | MEDLINE | ID: mdl-27777356

ABSTRACT

Chronic electric activation of the carotid baroreflex produces sustained reductions in sympathetic activity and arterial pressure and is currently being evaluated for therapy in patients with resistant hypertension. However, patients with significant impairment of renal function have been largely excluded from clinical trials. Thus, there is little information on blood pressure and renal responses to baroreflex activation in subjects with advanced chronic kidney disease, which is common in resistant hypertension. Changes in arterial pressure and glomerular filtration rate were determined in 5 dogs after combined unilateral nephrectomy and surgical excision of the poles of the remaining kidney to produce ≈70% reduction in renal mass. After control measurements, sodium intake was increased from ≈45 to 450 mol/d. While maintained on high salt, animals experienced increases in mean arterial pressure from 102±4 to 121±6 mm Hg and glomerular filtration rate from 40±2 to 45±2 mL/min. During 7 days of baroreflex activation, the hypertension induced by high salt was abolished (103±6 mm Hg) along with striking suppression of plasma norepinephrine concentration from 139±21 to 81±9 pg/mL, but despite pronounced blood pressure lowering, there were no significant changes in glomerular filtration rate (43±2 mL/min). All variables returned to prestimulation values during a recovery period. These findings indicate that after appreciable nephron loss, chronic suppression of central sympathetic outflow by baroreflex activation abolishes hypertension induced by high salt intake. The sustained antihypertensive effects of baroreflex activation occur without significantly compromising glomerular filtration rate in remnant nephrons.


Subject(s)
Baroreflex/physiology , Electric Stimulation , Hypertension/physiopathology , Sodium Chloride, Dietary/pharmacology , Animals , Baroreflex/drug effects , Blood Pressure Determination , Disease Models, Animal , Dogs , Glomerular Filtration Rate/physiology , Hypertension/chemically induced , Kidney/physiology , Male , Organ Size , Random Allocation , Reference Values , Risk Assessment
7.
Hypertension ; 68(1): 227-35, 2016 07.
Article in English | MEDLINE | ID: mdl-27160198

ABSTRACT

Carotid bodies play a critical role in protecting against hypoxemia, and their activation increases sympathetic activity, arterial pressure, and ventilation, responses opposed by acute stimulation of the baroreflex. Although chemoreceptor hypersensitivity is associated with sympathetically mediated hypertension, the mechanisms involved and their significance in the pathogenesis of hypertension remain unclear. We investigated the chronic interactions of these reflexes in dogs with sympathetically mediated, obesity-induced hypertension based on the hypothesis that hypoxemia and tonic activation of carotid chemoreceptors may be associated with obesity. After 5 weeks on a high-fat diet, the animals experienced a 35% to 40% weight gain and increases in arterial pressure from 106±3 to 123±3 mm Hg and respiratory rate from 8±1 to 12±1 breaths/min along with hypoxemia (arterial partial pressure of oxygen=81±3 mm Hg) but eucapnia. During 7 days of carotid baroreflex activation by electric stimulation of the carotid sinus, tachypnea was attenuated, and hypertension was abolished before these variables returned to prestimulation values during a recovery period. After subsequent denervation of the carotid sinus region, respiratory rate decreased transiently in association with further sustained reductions in arterial partial pressure of oxygen (to 65±2 mm Hg) and substantial hypercapnia. Moreover, the severity of hypertension was attenuated from 125±2 to 116±3 mm Hg (45%-50% reduction). These findings suggest that hypoxemia may account for sustained stimulation of peripheral chemoreceptors in obesity and that this activation leads to compensatory increases in ventilation and central sympathetic outflow that contributes to neurogenically mediated hypertension. Furthermore, the excitatory effects of chemoreceptor hyperactivity are abolished by chronic activation of the carotid baroreflex.


Subject(s)
Carotid Body , Hypertension/physiopathology , Obesity/physiopathology , Pressoreceptors/metabolism , Tachypnea/physiopathology , Animals , Chemoreceptor Cells/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Dogs , Electric Stimulation/methods , Hypertension/complications , Hypertension/therapy , Hypoxia/etiology , Hypoxia/physiopathology , Obesity/complications , Random Allocation , Tachypnea/etiology , Treatment Outcome
9.
Am J Physiol Renal Physiol ; 309(7): F583-94, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26224718

ABSTRACT

When introduced clinically 6 years ago, renal denervation was thought to be the solution for all patients whose blood pressure could not be controlled by medication. The initial two studies, SYMPLICITY HTN-1 and HTN-2, demonstrated great magnitudes of blood pressure reduction within 6 mo of the procedure and were based on a number of assumptions that may not have been true, including strict adherence to medication and absence of white-coat hypertension. The SYMPLICITY HTN-3 trial controlled for all possible factors believed to influence the outcome, including the addition of a sham arm, and ultimately proved the demise of the initial overly optimistic expectations. This trial yielded a much lower blood pressure reduction compared with the previous SYMPLICITY trials. Since its publication in 2014, there have been many analyses to try and understand what accounted for the differences. Of all the variables examined that could influence blood pressure outcomes, the extent of the denervation procedure was determined to be inadequate. Beyond this, the physiological mechanisms that account for the heterogeneous fall in arterial pressure following renal denervation remain unclear, and experimental studies indicate dependence on more than simply reduced renal sympathetic activity. These and other related issues are discussed in this paper. Our perspective is that renal denervation works if done properly and used in the appropriate patient population. New studies with new approaches and catheters and appropriate controls will be starting later this year to reassess the efficacy and safety of renal denervation in humans.


Subject(s)
Denervation/methods , Hypertension, Renal/surgery , Kidney/surgery , Drug Resistance , Humans , Hypertension, Renal/drug therapy , Renal Circulation , Sympathectomy
10.
Hypertension ; 65(6): 1223-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25895584

ABSTRACT

Recent technology for chronic electric activation of the carotid baroreflex and renal nerve ablation provide global and renal-specific suppression of sympathetic activity, respectively, but the conditions for favorable antihypertensive responses in resistant hypertension are unclear. Because inappropriately high plasma levels of aldosterone are prevalent in these patients, we investigated the effects of baroreflex activation and surgical renal denervation in dogs with hypertension induced by chronic infusion of aldosterone (12 µg/kg per day). Under control conditions, basal values for mean arterial pressure and plasma norepinephrine concentration were 100±3 mm Hg and 134±26 pg/mL, respectively. By day 7 of baroreflex activation, plasma norepinephrine was reduced by ≈40% and arterial pressure by 16±2 mm Hg. All values returned to control levels during the recovery period. Arterial pressure increased to 122±5 mm Hg concomitant with a rise in plasma aldosterone concentration from 4.3±0.4 to 70.0±6.4 ng/dL after 14 days of aldosterone infusion, with no significant effect on plasma norepinephrine. After 7 days of baroreflex activation at control stimulation parameters, the reduction in plasma norepinephrine was similar but the fall in arterial pressure (7±1 mm Hg) was diminished (≈55%) during aldosterone hypertension when compared with control conditions. Despite sustained suppression of sympathetic activity, baroreflex activation did not have central actions to inhibit either the stimulation of vasopressin secretion or drinking induced by increased plasma osmolality during chronic aldosterone infusion. Finally, renal denervation did not attenuate aldosterone hypertension. These findings suggest that aldosterone excess may portend diminished blood pressure lowering to global and especially renal-specific sympathoinhibition during device-based therapy.


Subject(s)
Aldosterone/pharmacology , Baroreflex/physiology , Hypertension/surgery , Norepinephrine/blood , Renin-Angiotensin System/drug effects , Analysis of Variance , Animals , Baroreflex/drug effects , Blood Pressure/physiology , Disease Models, Animal , Dogs , Enzyme-Linked Immunosorbent Assay , Hypertension/chemically induced , Hypertension/physiopathology , Linear Models , Male , Random Allocation , Reference Values , Renin/blood , Risk Assessment , Sensitivity and Specificity , Sympathectomy/methods
11.
Physiology (Bethesda) ; 30(2): 148-58, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25729060

ABSTRACT

Because of resetting, a role for baroreflexes in long-term control of arterial pressure has been commonly dismissed in the past. However, in recent years, this perspective has changed. Novel approaches for determining chronic neurohormonal and cardiovascular responses to natural variations in baroreceptor activity and to electrical stimulation of the carotid baroreflex indicate incomplete resetting and sustained responses that lead to long-term alterations in sympathetic activity and arterial pressure.


Subject(s)
Arterial Pressure , Baroreflex , Cardiovascular System/innervation , Mechanotransduction, Cellular , Pressoreceptors/physiology , Animals , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Baroreflex/drug effects , Homeostasis , Humans , Hypertension/drug therapy , Hypertension/etiology , Hypertension/physiopathology , Models, Cardiovascular , Obesity/complications , Obesity/physiopathology , Pressoreceptors/drug effects , Renin-Angiotensin System , Time Factors
12.
Curr Hypertens Rep ; 16(8): 453, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899538

ABSTRACT

Recent technical advances have led to the development of a medical device that can reliably activate the carotid baroreflex with an acceptable degree of safety. Because activation of the sympathetic nervous system plays an important role in the pathogenesis of hypertension and heart failure, the unique ability of this device to chronically suppress central sympathetic outflow in a controlled manner suggests potential value in the treatment of these conditions. This notion is supported by both clinical and experimental animal studies, and the major aim of this article is to elucidate the physiological mechanisms that account for the favorable effects of baroreflex activation therapy in patients with resistant hypertension and heart failure. Illumination of the neurohormonal, renal, and cardiac actions of baroreflex activation is likely to provide the means for better identification of those patients that are most likely to respond favorably to this device-based therapy.


Subject(s)
Baroreflex/physiology , Cardiovascular Diseases/therapy , Equipment and Supplies , Sympathetic Nervous System/physiopathology , Animals , Blood Pressure/physiology , Heart/innervation , Heart/physiology , Humans
13.
Hypertension ; 64(3): 604-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24935941

ABSTRACT

Chronic electric activation of the carotid baroreflex produces sustained reductions in sympathetic activity and arterial pressure and is currently being evaluated as antihypertensive therapy for patients with resistant hypertension. However, the influence of variations in salt intake on blood pressure lowering during baroreflex activation (BA) has not yet been determined. As the sensitivity of arterial pressure to salt intake is linked to the responsiveness of renin secretion, we determined steady-state levels of arterial pressure and neurohormonal responses in 6 dogs on low, normal, and high salt intakes (5, 40, 450 mmol/d, respectively) under control conditions and during a 7-day constant level of BA. Under control conditions, there was no difference in mean arterial pressure at low (92±1) and normal (92±2 mm Hg) sodium intakes, but pressure increased 9±2 mm Hg during high salt. Plasma renin activity (2.01±0.23, 0.93±0.20, 0.01±0.01 ng angiotensin I/mL/h) and plasma aldosterone (10.3±1.9, 3.5±0.5, 1.7±0.1 ng/dL) were inversely related to salt intake, whereas there were no changes in plasma norepinephrine. Although mean arterial pressure (19-22 mm Hg) and norepinephrine (20%-40%) were lower at all salt intakes during BA, neither the changes in pressure nor the absolute values for plasma renin activity or aldosterone in response to salt were different from control conditions. These findings demonstrate that suppression of sympathetic activity by BA lowers arterial pressure without increasing renin release and indicate that changes in sympathetic activity are not primary mediators of the effect of salt on renin secretion. Consequently, blood pressure lowering during BA is independent of salt intake.


Subject(s)
Baroreflex/physiology , Blood Pressure/physiology , Renin/blood , Sodium Chloride, Dietary/pharmacology , Aldosterone/blood , Animals , Blood Pressure/drug effects , Dogs , Dose-Response Relationship, Drug , Heart Rate/drug effects , Heart Rate/physiology , Hematocrit , Models, Animal , Neurotransmitter Agents/metabolism , Renin/metabolism
15.
Handb Clin Neurol ; 117: 395-406, 2013.
Article in English | MEDLINE | ID: mdl-24095142

ABSTRACT

Recent technical advances have renewed interest in device-based therapy for the treatment of drug-resistant hypertension. Findings from recent clinical trials regarding the efficacy of electrical stimulation of the carotid sinus for the treatment of resistant hypertension are reviewed here. However, a major goal of this article is to summarize experimental studies that have provided a conceptual understanding of the mechanisms that account for the long-term blood pressure lowering of arterial pressure with baroreflex activation. In so doing, the mechanistic insight from these studies may help to identify subsets of this heterogeneous population that stand to benefit the most. In this regard, because clinical, experimental, and theoretical evidence indicates that the kidneys play a dominant role in long-term control of arterial pressure, this article focuses on the mechanisms that link baroreflex-induced reductions in central sympathetic outflow with increases in renal excretory function that lead to sustained reductions in arterial pressure. Despite the encouraging findings from recent clinical trials, more basic research and additional clinical trials are needed to better define the benefit of baroreflex activation therapy in resistant hypertension and in other states of sympathetic activation such as heart failure and advanced renal disease.


Subject(s)
Autonomic Nervous System/physiology , Baroreflex/physiology , Hypertension/therapy , Animals , Humans , Hypertension/physiopathology
16.
Am J Physiol Heart Circ Physiol ; 305(7): H1080-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23913707

ABSTRACT

The sensitivity of baroreflex control of heart rate is depressed in subjects with obesity hypertension, which increases the risk for cardiac arrhythmias. The mechanisms are not fully known, and there are no therapies to improve this dysfunction. To determine the cardiovascular dynamic effects of progressive increases in body weight leading to obesity and hypertension in dogs fed a high-fat diet, 24-h continuous recordings of spontaneous fluctuations in blood pressure and heart rate were analyzed in the time and frequency domains. Furthermore, we investigated whether autonomic mechanisms stimulated by chronic baroreflex activation and renal denervation-current therapies in patients with resistant hypertension, who are commonly obese-restore cardiovascular dynamic control. Increases in body weight to ∼150% of control led to a gradual increase in mean arterial pressure to 17 ± 3 mmHg above control (100 ± 2 mmHg) after 4 wk on the high-fat diet. In contrast to the gradual increase in arterial pressure, tachycardia, attenuated chronotropic baroreflex responses, and reduced heart rate variability were manifest within 1-4 days on high-fat intake, reaching 130 ± 4 beats per minute (bpm) (control = 86 ± 3 bpm) and ∼45% and <20%, respectively, of control levels. Subsequently, both baroreflex activation and renal denervation abolished the hypertension. However, only baroreflex activation effectively attenuated the tachycardia and restored cardiac baroreflex sensitivity and heart rate variability. These findings suggest that baroreflex activation therapy may reduce the risk factors for cardiac arrhythmias as well as lower arterial pressure.


Subject(s)
Baroreflex , Electric Stimulation Therapy , Heart Rate , Hypertension/therapy , Kidney/innervation , Obesity/complications , Sympathectomy/methods , Tachycardia/prevention & control , Animals , Arterial Pressure , Disease Models, Animal , Dogs , Hypertension/etiology , Hypertension/physiopathology , Male , Obesity/physiopathology , Tachycardia/etiology , Tachycardia/physiopathology , Time Factors
17.
Curr Hypertens Rep ; 15(4): 409-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23677623

ABSTRACT

Abundant evidence supports a role of the sympathetic nervous system in the pathogenesis of obesity-related hypertension. However, the nature and temporal progression of mechanisms underlying this sympathetically mediated hypertension are incompletely understood. Recent technological advances allowing direct recordings of renal sympathetic nerve activity (RSNA) in conscious animals, together with direct suppression of RSNA by renal denervation and reflex-mediated global sympathetic inhibition in experimental animals and human subjects have been especially valuable in elucidating these mechanisms. These studies strongly support the concept that increased RSNA is the critical mechanism by which increased central sympathetic outflow initiates and maintains reductions in renal excretory function, causing obesity hypertension. Potential determinants of renal sympathoexcitation and the differential mechanisms mediating the effects of renal-specific versus reflex-mediated, global sympathetic inhibition on renal hemodynamics and cardiac autonomic function are discussed. These differential mechanisms may impact the efficacy of current device-based approaches for hypertension therapy.


Subject(s)
Hypertension/physiopathology , Obesity/physiopathology , Renin-Angiotensin System/physiology , Sympathetic Nervous System/physiopathology , Animals , Denervation/methods , Humans , Hypertension/etiology , Kidney/physiopathology , Obesity/complications , Sympathetic Nervous System/drug effects
18.
Hypertension ; 60(3): 749-56, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22753216

ABSTRACT

Chronic electric activation of the carotid baroreflex produces sustained reductions in sympathetic activity and arterial pressure and is currently being evaluated as hypertension therapy for patients with resistant hypertension. However, the chronic changes in renal function associated with natural suppression of sympathetic activity are largely unknown. In normotensive dogs, we investigated the integrative cardiovascular effects of chronic baroreflex activation (2 weeks) alone and in combination with the calcium channel blocker amlodipine, which is commonly used in the treatment of resistant hypertension. During baroreflex activation alone, there were sustained decreases in mean arterial pressure (17±1 mmHg) and plasma (norepinephrine; ≈35%), with no change in plasma renin activity. Despite low pressure, sodium balance was achieved because of decreased tubular reabsorption, because glomerular filtration rate and renal blood flow decreased 10% to 20%. After 2 weeks of amlodipine, arterial pressure was also reduced 17 mmHg, but with substantial increases in norepinephrine and plasma renin activity and no change in glomerular filtration rate. In the presence of amlodipine, baroreflex activation greatly attenuated neurohormonal activation, and pressure decreased even further (by 11±2 mmHg). Moreover, during amlodipine administration, the fall in glomerular filtration rate with baroreflex activation was abolished. These findings suggest that the chronic blood pressure-lowering effects of baroreflex activation are attributed, at least in part, to sustained inhibition of renal sympathetic nerve activity and attendant decreases in sodium reabsorption before the macula densa. Tubuloglomerular feedback constriction of the afferent arterioles may account for reduced glomerular filtration rate, a response abolished by amlodipine, which dilates the preglomerular vasculature.


Subject(s)
Baroreflex/physiology , Carotid Arteries/physiology , Kidney/blood supply , Kidney/physiology , Regional Blood Flow/physiology , Sympathetic Nervous System/physiology , Amlodipine/pharmacology , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Calcium Channel Blockers/pharmacology , Dogs , Electric Stimulation , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Kidney/innervation , Models, Animal , Norepinephrine/blood , Regional Blood Flow/drug effects , Renin/blood , Sodium/metabolism
19.
J Appl Physiol (1985) ; 113(10): 1652-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22797307

ABSTRACT

Device-based therapy for resistant hypertension by electrical activation of the carotid baroreflex is currently undergoing active clinical investigation, and initial findings from clinical trials have been published. The purpose of this mini-review is to summarize the experimental studies that have provided a conceptual understanding of the mechanisms that account for the long-term lowering of arterial pressure with baroreflex activation. The well established mechanisms mediating the role of the baroreflex in short-term regulation of arterial pressure by rapid changes in peripheral resistance and cardiac function are often extended to long-term pressure control, and the more sluggish actions of the baroreflex on renal excretory function are often not taken into consideration. However, because clinical, experimental, and theoretical evidence indicates that the kidneys play a dominant role in long-term control of arterial pressure, this review focuses on the mechanisms that link baroreflex-mediated reductions in central sympathetic outflow with increases in renal excretory function that lead to sustained reductions in arterial pressure.


Subject(s)
Arterial Pressure , Baroreflex , Carotid Sinus/innervation , Central Nervous System/physiopathology , Electric Stimulation Therapy , Hypertension/therapy , Kidney/innervation , Sympathetic Nervous System/physiopathology , Animals , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Drug Resistance , Humans , Hypertension/etiology , Hypertension/metabolism , Hypertension/physiopathology , Obesity/complications , Renin-Angiotensin System , Treatment Outcome , Vascular Resistance
SELECTION OF CITATIONS
SEARCH DETAIL
...