Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Kidney Dis ; 80(4): 527-535, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35598810

RESUMO

Uncontrolled hypertension persists as an important health issue despite the availability of many medications and nondrug therapies that lower blood pressure. Increasingly, nonadherence to medication is found in approximately 2 of every 5 patients with uncontrolled hypertension. In the search for interventions that lower blood pressure that do not rely on adherence to a regimen requiring daily ingestion of medication or repeated physical activity, device-based methods that denervate the renal arteries have emerged as a potential complement to standard antihypertensive treatments. At least 3 different approaches to renal artery denervation are under active investigation, including the use of radiofrequency energy, ultrasound, or the injection of neurolytic agents into the renal perivascular tissue. In this review, we cover what is currently known about the mechanisms of antihypertensive effects of renal denervation, summarize the efficacy and safety of renal denervation using recent controlled trial publications in a number of hypertensive populations, and conclude with some thoughts about challenges in the field, including the optimization of patient selection for the procedure and what the reader can expect in the near future in this rapidly developing field.


Assuntos
Anti-Hipertensivos , Hipertensão , Anti-Hipertensivos/uso terapêutico , Pressão Sanguínea , Denervação/métodos , Humanos , Hipertensão/tratamento farmacológico , Rim , Artéria Renal , Simpatectomia/métodos , Resultado do Tratamento
2.
Front Physiol ; 12: 771167, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34916958

RESUMO

We have previously reported that brain Gαi2 subunit proteins are required to maintain sodium homeostasis and are endogenously upregulated in the hypothalamic paraventricular nucleus (PVN) in response to increased dietary salt intake to maintain a salt resistant phenotype in rats. However, the origin of the signal that drives the endogenous activation and up-regulation of PVN Gαi2 subunit protein signal transduction pathways is unknown. By central oligodeoxynucleotide (ODN) administration we show that the pressor responses to central acute administration and central infusion of sodium chloride occur independently of brain Gαi2 protein pathways. In response to an acute volume expansion, we demonstrate, via the use of selective afferent renal denervation (ADNX) and anteroventral third ventricle (AV3V) lesions, that the sensory afferent renal nerves, but not the sodium sensitive AV3V region, are mechanistically involved in Gαi2 protein mediated natriuresis to an acute volume expansion [peak natriuresis (µeq/min) sham AV3V: 43 ± 4 vs. AV3V 45 ± 4 vs. AV3V + Gαi2 ODN 25 ± 4, p < 0.05; sham ADNX: 43 ± 4 vs. ADNX 23 ± 6, AV3V + Gαi2 ODN 25 ± 3, p < 0.05]. Furthermore, in response to chronically elevated dietary sodium intake, endogenous up-regulation of PVN specific Gαi2 proteins does not involve the AV3V region and is mediated by the sensory afferent renal nerves to counter the development of the salt sensitivity of blood pressure (MAP [mmHg] 4% NaCl; Sham ADNX 124 ± 4 vs. ADNX 145 ± 4, p < 0.05; Sham AV3V 125 ± 4 vs. AV3V 121 ± 5). Additionally, the development of the salt sensitivity of blood pressure following central ODN-mediated Gαi2 protein down-regulation occurs independently of the actions of the brain angiotensin II type 1 receptor. Collectively, our data suggest that in response to alterations in whole body sodium the peripheral sensory afferent renal nerves, but not the central AV3V sodium sensitive region, evoke the up-regulation and activation of PVN Gαi2 protein gated pathways to maintain a salt resistant phenotype. As such, both the sensory afferent renal nerves and PVN Gαi2 protein gated pathways, represent potential targets for the treatment of the salt sensitivity of blood pressure.

3.
Annu Rev Physiol ; 83: 429-450, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33566672

RESUMO

Renal sympathetic (efferent) nerves play an important role in the regulation of renal function, including glomerular filtration, sodium reabsorption, and renin release. The kidney is also innervated by sensory (afferent) nerves that relay information to the brain to modulate sympathetic outflow. Hypertension and other cardiometabolic diseases are linked to overactivity of renal sympathetic and sensory nerves, but our mechanistic understanding of these relationships is limited. Clinical trials of catheter-based renal nerve ablation to treat hypertension have yielded promising results. Therefore, a greater understanding of how renal nerves control the kidney under physiological and pathophysiological conditions is needed. In this review, we provide an overview of the current knowledge of the anatomy of efferent and afferent renal nerves and their functions in normal and pathophysiological conditions. We also suggest further avenues of research for development of novel therapies targeting the renal nerves.


Assuntos
Vias Aferentes/fisiologia , Hipertensão/fisiopatologia , Rim/inervação , Rim/fisiologia , Animais , Ablação por Cateter/métodos , Humanos , Rim/fisiopatologia
4.
J Clin Med Res ; 13(12): 556-562, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35059074

RESUMO

BACKGROUND: No methods exist for confirming nerve ablation in catheter-based renal sympathetic denervation (RDN). METHODS: We investigated the feasibility of using intravascular ultrasound (IVUS) to locate nerves and observe nerve integrity changes during RDN in a pig. To confirm our observations, we used post-RDN histological sections matched anatomically to the IVUS images. RESULTS: IVUS revealed multiple hypoechoic structures along the renal artery, whose locations matched those of nerves in the histological sections. Nerves clustered near the junction between the renal artery and vein. Histology confirmed necrosis of nerve bundles at RDN ablation sites, but no changes in echogenicity were observed using IVUS. CONCLUSIONS: Although IVUS cannot currently be used to confirm ablation during RDN, it clearly reveals some clusters of renal sympathetic nerves. It remains to be demonstrated how IVUS can guide RDN devices and potentially improve ablation success.

5.
Front Physiol ; 12: 784957, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35111076

RESUMO

The mechanisms that sense alterations in total body sodium content to facilitate sodium homeostasis in response to an acute sodium challenge that does not increase blood pressure have not been fully elucidated. We hypothesized that the renal sympathetic nerves are critical to mediate natriuresis via α1- or ß-adrenoceptors signal transduction pathways to maintain sodium balance in the face of acute increases in total body sodium content that do not activate the pressure-natriuresis mechanism. To address this hypothesis, we used acute bilateral renal denervation (RDNX), an anteroventral third ventricle (AV3V) lesion and α1- or ß-antagonism during an acute 1M NaCl sodium challenge in conscious male Sprague Dawley rats. An acute 1M NaCl infusion did not alter blood pressure and evoked profound natriuresis and sympathoinhibition. Acute bilateral RDNX attenuated the natriuretic and sympathoinhibitory responses evoked by a 1M NaCl infusion [peak natriuresis (µeq/min) sham 14.5 ± 1.3 vs. acute RDNX: 9.2 ± 1.4, p < 0.05; plasma NE (nmol/L) sham control: 44 ± 4 vs. sham 1M NaCl infusion 11 ± 2, p < 0.05; acute RDNX control: 42 ± 6 vs. acute RDNX 1M NaCl infusion 25 ± 3, p < 0.05]. In contrast, an AV3V lesion did not impact the cardiovascular, renal excretory or sympathoinhibitory responses to an acute 1M NaCl infusion. Acute i.v. α1-adrenoceptor antagonism with terazosin evoked a significant drop in baseline blood pressure and significantly attenuated the natriuretic response to a 1M NaCl load [peak natriuresis (µeq/min) saline 17.2 ± 1.4 vs. i.v. terazosin 7.8 ± 2.5, p < 0.05]. In contrast, acute ß-adrenoceptor antagonism with i.v. propranolol infusion did not impact the cardiovascular or renal excretory responses to an acute 1M NaCl infusion. Critically, the natriuretic response to an acute 1M NaCl infusion was significantly blunted in rats receiving a s.c. infusion of the α1-adrenoceptor antagonist terazosin at a dose that did not lower baseline blood pressure [peak natriuresis (µeq/min) sc saline: 18 ± 1 vs. sc terazosin 7 ± 2, p < 0.05]. Additionally, a s.c. infusion of the α1-adrenoceptor antagonist terazosin further attenuated the natriuretic response to a 1M NaCl infusion in acutely RDNX animals. Collectively these data indicate a specific role of a blood pressure-independent renal sympathetic nerve-dependent α1-adrenoceptor-mediated pathway in the natriuretic and sympathoinhibitory responses evoked by acute increases in total body sodium.

7.
Front Physiol ; 11: 725, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32670095

RESUMO

Children born with a solitary functioning kidney (SFK) have an increased risk of hypertension and kidney disease from early in adulthood. In response to a reduction in kidney mass, the remaining kidney undergoes compensatory kidney growth. This is associated with both an increase in size of the kidney tubules and the glomeruli and an increase in single nephron glomerular filtration rate (SNGFR). The compensatory hypertrophy and increase in filtration at the level of the individual nephron results in normalization of total glomerular filtration rate (GFR). However, over time these same compensatory mechanisms may contribute to kidney injury and hypertension. Indeed, approximately 50% of children born with a SFK develop hypertension by the age of 18 and 20-40% require dialysis by the age of 30. The mechanisms that result in kidney injury are only partly understood, and early biomarkers that distinguish those at an elevated risk of kidney injury are needed. This review will outline the compensatory adaptations to a SFK, and outline how these adaptations may contribute to kidney injury and hypertension later in life. These will be based largely on the mechanisms we have identified from our studies in an ovine model of SFK, that implicate the renal nitric oxide system, the renin angiotensin system and the renal nerves to kidney disease and hypertension associated with SFK. This discussion will also evaluate current, and speculate on next generation, prognostic factors that may predict those children at a higher risk of future kidney disease and hypertension.

8.
Hypertension ; 75(4): 1002-1011, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32148128

RESUMO

We have previously reported that in salt-resistant rat phenotypes brain, Gαi2 (guanine nucleotide-binding protein alpha inhibiting activity polypeptide 2) proteins are required to maintain blood pressure and sodium balance. However, the impact of hypothalamic paraventricular nucleus (PVN) Gαi2 proteins on the salt sensitivity of blood pressure is unknown. Here, by the bilateral PVN administration of a targeted Gαi2 oligodeoxynucleotide, we show that PVN-specific Gαi2 proteins are required to facilitate the full natriuretic response to an acute volume expansion (peak natriuresis [µeq/min] scrambled (SCR) oligodeoxynucleotide 41±3 versus Gαi2 oligodeoxynucleotide 18±4; P<0.05) via a renal nerve-dependent mechanism. Furthermore, in response to chronically elevated dietary sodium intake, PVN-specific Gαi2 proteins are essential to counter renal nerve-dependent salt-sensitive hypertension (mean arterial pressure [mm Hg] 8% NaCl; SCR oligodeoxynucleotide 128±2 versus Gαi2 oligodeoxynucleotide 147±3; P<0.05). This protective pathway involves activation of PVN Gαi2 signaling pathways, which mediate sympathoinhibition to the blood vessels and kidneys (renal norepinephrine [pg/mg] 8% NaCl; SCR oligodeoxynucleotide 375±39 versus Gαi2 oligodeoxynucleotide 850±27; P<0.05) and suppression of the activity of the sodium chloride cotransporter assessed as peak natriuresis to hydrochlorothiazide. Additionally, central oligodeoxynucleotide-mediated Gαi2 protein downregulation prevented PVN parvocellular neuron activation, assessed by FosB immunohistochemistry, in response to increased dietary salt intake. In our analysis of the UK BioBank data set, it was observed that 2 GNAI2 single nucleotide polymorphism (SNP) (rs2298952, P=0.041; rs4547694, P=0.017) significantly correlate with essential hypertension. Collectively, our data suggest that selective targeting and activation of PVN Gαi2 proteins is a novel therapeutic approach for the treatment of salt-sensitive hypertension.


Assuntos
Pressão Sanguínea/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Hipertensão/metabolismo , Rim/metabolismo , Natriurese/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Cloreto de Sódio na Dieta , Animais , Masculino , Vias Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
9.
Clin Sci (Lond) ; 133(20): 2061-2067, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31654065

RESUMO

The precise mechanisms underlying resistant hypertension remain elusive. Reduced nitric oxide (NO) bioavailability is frequently documented in chronic kidney disease, obesity, diabetes and advanced age, all of which are risk factors for resistant hypertension. Sympathetic overactivity and chronic activation of the renin-angiotensin system are salient features of resistant hypertension. Interestingly, recent data indicate that renal sympathetic overactivity can reduce the expression of neuronal nitric oxide synthase in the paraventricular nucleus. Reduced NO levels in the paraventricular nucleus can increase sympathetic outflow and this can create a vicious cycle contributing to resistant hypertension. Angiotensin II can reduce l-arginine transport and hence NO production. Reduced NO levels may reduce the formation of angiotensin 1-7 dampening the cardio-protective effects of the renin-angiotensin system contributing to resistant hypertension. In addition, interleukin-6 (IL-6) is demonstrated to be independently associated with resistant hypertension, and IL-6 can reduce NO synthesis. Despite this, NO levels have not been quantified in resistant hypertension. Findings from a small proof of concept study indicate that NO donors can reduce blood pressure in patients with resistant hypertension but more studies are required to validate these preliminary findings. In the present paper, we put forward the hypothesis that reduced NO bioavailability contributes substantially to the development of resistant hypertension.


Assuntos
Arginina/fisiologia , Hipertensão/fisiopatologia , Óxido Nítrico/fisiologia , Disponibilidade Biológica , Endotélio Vascular/fisiopatologia , Humanos , Hipertensão/etiologia , Hipertensão/terapia , Inflamação/complicações , Óxido Nítrico/deficiência , Óxido Nítrico/farmacocinética , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Falha de Tratamento , Rigidez Vascular/fisiologia
10.
11.
Heart Rhythm ; 14(6): 912-919, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28193508

RESUMO

BACKGROUND: Our previous studies showed that renal sympathetic stimulation (RS) may facilitate ischemic ventricular arrhythmia (VA) by increasing left stellate ganglion (LSG) nerve activity, whereas renal sympathetic ablation (RA) may suppress VA. OBJECTIVE: The purpose of this study was to investigate whether renal sympathetic interventions also can affect VA by modulating LSG activity in a cesium-induced long QT canine model. METHODS: Twenty-four dogs were randomly divided into RS group (n = 8), RA group (n = 8), or control group (n = 8). Serum norepinephrine, LSG function, and LSG neural activity were measured before and 3 hours after RS or RA. Increasing doses of cesium chloride then were administered until a "threshold dose" produced sustained ventricular tachycardia or ventricular fibrillation. Early afterdepolarization amplitude, VA prevalence, and tachycardia threshold dose were compared among these groups. Nerve growth factor and c-fos protein expressed in the LSG also were examined. RESULTS: Serum norepinephrine, LSG function, and LSG neural activity were all significantly increased after 3 hours of RS and all were decreased 3 hours after RA. In addition, RS significantly decreased the tachycardia threshold dose, increased the early afterdepolarization amplitude, facilitated the incidence of VAs, and increased the expression of nerve growth factor and c-fos protein. In contrast, RA induced the opposite effects. CONCLUSION: RS promotes, whereas RA suppresses, the incidence of VAs in a canine model of cesium-induced long QT. Modulation of LSG neural activity by RS and RA may be responsible for these different effects.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Ablação por Cateter/métodos , Terapia por Estimulação Elétrica/métodos , Eletrocardiografia , Frequência Cardíaca/fisiologia , Rim/inervação , Taquicardia Ventricular/terapia , Animais , Sistema Nervoso Autônomo/cirurgia , Césio/toxicidade , Modelos Animais de Doenças , Cães , Masculino , Gânglio Estrelado/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Sistema Nervoso Simpático/cirurgia , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/fisiopatologia
12.
Acta Physiol (Oxf) ; 220(4): 404-416, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28181735

RESUMO

Extensive investigations have revealed that renal sympathetic nerves regulate renin secretion, tubular fluid reabsorption and renal haemodynamics which can impact on cardiovascular homoeostasis normally and in pathophysiological states. The significance of the renal afferent innervation and its role in determining the autonomic control of the cardiovascular system is uncertain. The transduction pathways at the renal afferent nerves have been shown to require pro-inflammatory mediators and TRPV1 channels. Reno-renal reflexes have been described, both inhibitory and excitatory, demonstrating that a neural link exists between kidneys and may determine the distribution of excretory and haemodynamic function between the two kidneys. The impact of renal afferent nerve activity on basal and reflex regulation of global sympathetic drive remains opaque. There is clinical and experimental evidence that in states of chronic kidney disease and renal injury, there is infiltration of T-helper cells with a sympatho-excitation and blunting of the high- and low-pressure baroreceptor reflexes regulating renal sympathetic nerve activity. The baroreceptor deficits are renal nerve-dependent as the dysregulation can be relieved by renal denervation. There is also experimental evidence that in obese states, there is a sympatho-excitation and disrupted baroreflex regulation of renal sympathetic nerve activity which is mediated by the renal innervation. This body of information provides an important basis for directing greater attention to the role of renal injury/inflammation causing an inappropriate activation of the renal afferent nerves as an important initiator of aberrant autonomic cardiovascular control.


Assuntos
Injúria Renal Aguda/fisiopatologia , Hemodinâmica/fisiologia , Inflamação/fisiopatologia , Rim/inervação , Sistema Nervoso Simpático/fisiologia , Animais , Sistema Cardiovascular/fisiopatologia , Humanos
13.
Am J Physiol Renal Physiol ; 310(1): F43-56, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26538440

RESUMO

Hydronephrosis is associated with the development of salt-sensitive hypertension. Studies have suggested that increased sympathetic nerve activity and oxidative stress play important roles in hypertension and the modulation of salt sensitivity. The present study primarily aimed to examine the role of renal sympathetic nerve activity in the development of hypertension in rats with hydronephrosis. In addition, we aimed to investigate if NADPH oxidase (NOX) function could be affected by renal denervation. Partial unilateral ureteral obstruction (PUUO) was created in 3-wk-old rats to induce hydronephrosis. Sham surgery or renal denervation was performed at the same time. Blood pressure was measured during normal, high-, and low-salt diets. The renal excretion pattern, NOX activity, and expression as well as components of the renin-angiotensin-aldosterone system were characterized after treatment with the normal salt diet. On the normal salt diet, rats in the PUUO group had elevated blood pressure compared with control rats (115 ± 3 vs. 87 ± 1 mmHg, P < 0.05) and displayed increased urine production and lower urine osmolality. The blood pressure change in response to salt loading (salt sensitivity) was more pronounced in the PUUO group compared with the control group (15 ± 2 vs. 5 ± 1 mmHg, P < 0.05). Renal denervation in PUUO rats attenuated both hypertension (97 ± 3 mmHg) and salt sensitivity (5 ± 1 mmHg, P < 0.05) and normalized the renal excretion pattern, whereas the degree of renal fibrosis and inflammation was not changed. NOX activity and expression as well as renin and ANG II type 1A receptor expression were increased in the renal cortex from PUUO rats and normalized by denervation. Plasma Na(+) and K(+) levels were elevated in PUUO rats and normalized after renal denervation. Finally, denervation in PUUO rats was also associated with reduced NOX expression, superoxide production, and fibrosis in the heart. In conclusion, renal denervation attenuates hypertension and restores the renal excretion pattern, which is associated with reduced renal NOX and components of the renin-angiotensin-aldosterone system. This study emphasizes a link between renal nerves, the development of hypertension, and modulation of NOX function.


Assuntos
Pressão Sanguínea , Hidronefrose/enzimologia , Hipertensão/cirurgia , Rim/enzimologia , Rim/inervação , NADPH Oxidases/metabolismo , Estresse Oxidativo , Simpatectomia/métodos , Animais , Biomarcadores/sangue , Biomarcadores/urina , Modelos Animais de Doenças , Fibrose , Frequência Cardíaca , Hidronefrose/etiologia , Hidronefrose/genética , Hidronefrose/fisiopatologia , Hipertensão/enzimologia , Hipertensão/etnologia , Hipertensão/genética , Hipertensão/fisiopatologia , Rim/patologia , Masculino , Miocárdio/enzimologia , Miocárdio/patologia , NADPH Oxidases/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Eliminação Renal , Sistema Renina-Angiotensina , Cloreto de Sódio na Dieta , Obstrução Ureteral/complicações
14.
Front Physiol ; 6: 233, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26347659

RESUMO

To counter the development of salt-sensitive hypertension, multiple brain G-protein-coupled receptor (GPCR) systems are activated to facilitate sympathoinhibition, sodium homeostasis, and normotension. Currently there is a paucity of knowledge regarding the role of down-stream GPCR-activated Gα-subunit proteins in these critically important physiological regulatory responses required for long-term blood pressure regulation. We have determined that brain Gαi2-proteins mediate natriuretic and sympathoinhibitory responses produced by acute pharmacological (exogenous central nociceptin/orphanin FQ receptor (NOP) and α2-adrenoceptor activation) and physiological challenges to sodium homeostasis (intravenous volume expansion and 1 M sodium load) in conscious Sprague-Dawley rats. We have demonstrated that in salt-resistant rat phenotypes, high dietary salt intake evokes site-specific up-regulation of hypothalamic paraventricular nucleus (PVN) Gαi2-proteins. Further, we established that PVN Gαi2 protein up-regulation prevents the development of renal nerve-dependent sympathetically mediated salt-sensitive hypertension in Sprague-Dawley and Dahl salt-resistant rats. Additionally, failure to up-regulate PVN Gαi2 proteins during high salt-intake contributes to the pathophysiology of Dahl salt-sensitive (DSS) hypertension. Collectively, our data demonstrate that brain, and likely PVN specific, Gαi2 protein pathways represent a central molecular pathway mediating sympathoinhibitory renal-nerve dependent responses evoked to maintain sodium homeostasis and a salt-resistant phenotype. Further, impairment of this endogenous "anti-hypertensive" mechanism contributes to the pathophysiology of salt-sensitive hypertension.

15.
Front Physiol ; 6: 224, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26300788

RESUMO

The function of the renal nerves has been an area of scientific and medical interest for many years. The recent advent of a minimally invasive catheter-based method of renal denervation has renewed excitement in understanding the afferent and efferent actions of the renal nerves in multiple diseases. While hypertension has been the focus of much this work, less attention has been given to the role of the renal nerves in the development of chronic heart failure (CHF). Recent studies from our laboratory and those of others implicate an essential role for the renal nerves in the development and progression of CHF. Using a rabbit tachycardia model of CHF and surgical unilateral renal denervation, we provide evidence for both renal efferent and afferent mechanisms in the pathogenesis of CHF. Renal denervation prevented the decrease in renal blood flow observed in CHF while also preventing increases in Angiotensin-II receptor protein in the microvasculature of the renal cortex. Renal denervation in CHF also reduced physiological markers of autonomic dysfunction including an improvement in arterial baroreflex function, heart rate variability, and decreased resting cardiac sympathetic tone. Taken together, the renal sympathetic nerves are necessary in the pathogenesis of CHF via both efferent and afferent mechanisms. Additional investigation is warranted to fully understand the role of these nerves and their role as a therapeutic target in CHF.

16.
Acta Physiol (Oxf) ; 213(3): 731-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25369876

RESUMO

AIMS: The renal sensory nerves are importantly involved in the sympathetic regulation of cardiovascular and renal function. Two reno-renal reflexes are recognized, one in which activation of renal sensory nerves elicits a renal sympatho-inhibition, and one which causes a renal sympatho-excitation and about which little is known. This study investigated the role of bradykinin (BK) in engaging an excitatory reno-renal reflex. METHODS: Rats were anaesthetized (chloralose/urethane) and prepared for the measurement of renal function or renal sympathetic nerve activity (RSNA). BK was infused into the cortico-medullary border of the ipsilateral kidney and the impact on contralateral renal function and RSNA evaluated. RESULTS: Intrarenal infusion of BK at 3 × 10(-9) and 6 × 10(-9) g L(-1) had no effect on mean arterial pressure, at 104 ± 5 mmHg or glomerular filtration rate in either the ipsilateral or contralateral kidneys, at 4.31 ± 0.45 mL min(-1) kg(-1) . At the highest dose of BK, fractional sodium excretion (FENa) was 1.47% in the ipsilateral kidney and was significantly lower, at 0.64% (P < 0.05) in the contralateral kidney but this difference did not occur following ipsilateral renal denervation. Ipsilateral intrarenal infusion of BK at 3 × 10(-9) , 6 × 10(-9) and 1.2 × 10(-8) g L(-1) elicited dose-related increases (P < 0.05) in contralateral RSNA, reaching some 78% at the highest dose, but these responses were prevented by ipsilateral renal denervation. CONCLUSIONS: Intrarenal infusion of BK produced an excitatory reno-renal reflex which was expressed as a renal nerve-dependent antinatriuresis in the contralateral kidney. The findings suggest that inflammatory mediators such as BK may be important in initiating a sympatho-excitation associated with renal and cardiovascular diseases.


Assuntos
Bradicinina/administração & dosagem , Mediadores da Inflamação/administração & dosagem , Rim/efeitos dos fármacos , Rim/inervação , Natriurese/efeitos dos fármacos , Reflexo/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Desequilíbrio Hidroeletrolítico/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Ratos Wistar , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais , Sistema Nervoso Simpático/fisiopatologia , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/fisiopatologia
17.
Hypertension ; 65(1): 178-86, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312437

RESUMO

Excess dietary salt intake is an established cause of hypertension. At present, our understanding of the neuropathophysiology of salt-sensitive hypertension is limited by a lack of identification of the central nervous system mechanisms that modulate sympathetic outflow and blood pressure in response to dietary salt intake. We hypothesized that impairment of brain Gαi2-protein-gated signal transduction pathways would result in increased sympathetically mediated renal sodium retention, thus promoting the development of salt-sensitive hypertension. To test this hypothesis, naive or renal denervated Dahl salt-resistant and Dahl salt-sensitive (DSS) rats were assigned to receive a continuous intracerebroventricular control scrambled or a targeted Gαi2-oligodeoxynucleotide infusion, and naive Brown Norway and 8-congenic DSS rats were fed a 21-day normal or high-salt diet. High salt intake did not alter blood pressure, suppressed plasma norepinephrine, and evoked a site-specific increase in hypothalamic paraventricular nucleus Gαi2-protein levels in naive Brown Norway, Dahl salt-resistant, and scrambled oligodeoxynucleotide-infused Dahl salt-resistant but not DSS rats. In Dahl salt-resistant rats, Gαi2 downregulation evoked rapid renal nerve-dependent hypertension, sodium retention, and sympathoexcitation. In DSS rats, Gαi2 downregulation exacerbated salt-sensitive hypertension via a renal nerve-dependent mechanism. Congenic-8 DSS rats exhibited sodium-evoked paraventricular nucleus-specific Gαi2-protein upregulation and attenuated hypertension, sodium retention, and global sympathoexcitation compared with DSS rats. These data demonstrate that paraventricular nucleus Gαi2-protein-gated pathways represent a conserved central molecular pathway mediating sympathoinhibitory renal nerve-dependent responses evoked to maintain sodium homeostasis and a salt-resistant phenotype. Impairment of this mechanism contributes to the development of salt-sensitive hypertension.


Assuntos
Pressão Sanguínea/fisiologia , Sistema Nervoso Central/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/biossíntese , Hipertensão/metabolismo , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Hipertensão/fisiopatologia , Immunoblotting , Masculino , Ratos , Ratos Endogâmicos Dahl , Transdução de Sinais , Sódio na Dieta/toxicidade
18.
J Cardiovasc Electrophysiol ; 25(11): 1249-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25066536

RESUMO

INTRODUCTION: Renal sympathetic nerve (RSN) activity plays a key role in systemic sympathetic hyperactivity. Previous studies have shown that cardiac sympathetic hyperactivity, especially the left stellate ganglion (LSG), contributes to the pathogenesis of ventricular arrhythmias (VAs) after acute myocardial infarction (AMI). METHODS AND RESULTS: Twenty-eight dogs received 3 hours of continuous left-sided electrical stimulation of RSN (LRS; Group-1, n = 9), sham RSN stimulation (Group-2, n = 9), or LSG ablation plus 3 hours of LRS (Group-3, n = 10) were included. AMI was induced by ligating the proximal left anterior descending coronary artery. LRS was performed using electrical stimulation on the adventitia of left renal artery at the voltage increasing the systolic blood pressure (BP) by 10%. BP, heart rate variability (HRV), serum norepinephrine (NE) level, and LSG function were measured at baseline and the end of each hour of LRS. C-fos and nerve growth factor (NGF) protein expressed in the LSG were examined in Group-1 and Group-2. Compared with baseline, 3 hours of LRS induced a significant increase in BP, sympathetic indices of HRV, serum NE level, and LSG function. The incidence of VAs in Group-1 was significantly higher than other groups. The expression of c-fos and NGF protein in the LSG was significantly higher in Group-1 than Group-2. CONCLUSION: Three hours of LRS induces both systemic and cardiac sympathetic hyperactivity and increases the incidence of ischemia-induced VAs.


Assuntos
Rim/inervação , Isquemia Miocárdica/fisiopatologia , Gânglio Estrelado/fisiologia , Taquicardia Ventricular/fisiopatologia , Animais , Cães , Estimulação Elétrica/efeitos adversos , Estimulação Elétrica/métodos , Rim/fisiologia , Masculino , Isquemia Miocárdica/complicações , Taquicardia Ventricular/etiologia
19.
Handb Clin Neurol ; 117: 203-14, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24095127

RESUMO

The kidneys play a central role in cardiovascular homeostasis by ensuring a balance between the fluid taken in and that lost and excreted during everyday activities. This ensures stability of extracellular fluid volume and maintenance of normal levels of blood pressure. Renal fluid handling is controlled via neural and humoral influences, with the former determining a rapid dynamic response to changing intake of sodium whereas the latter cause a slower longer-term modulation of sodium and water handling. Activity in the renal sympathetic nerves arises from an integration of information from the high and low pressure cardiovascular baroreceptors, the somatosensory and visceral systems as well as the higher cortical centers. Each sensory system provides varying input to the autonomic centers of the hypothalamic and medullary areas of the brain at a level appropriate to the activity being performed. In pathophysiological states, such as hypertension, heart failure and chronic renal disease, there may be an inappropriate sympathoexcitation causing sodium retention which exacerbates the disease process. The contribution of the renal sympathetic nerves to these cardiovascular diseases is beginning to be appreciated with the demonstration that renal denervation of resistant hypertensive patients results in a long-term normalization of blood pressure.


Assuntos
Vias Autônomas/fisiologia , Rim/fisiologia , Animais , Vias Autônomas/anatomia & histologia , Encéfalo/fisiologia , Humanos , Nefropatias/patologia , Nefropatias/fisiopatologia , Vias Neurais/fisiologia , Renina/metabolismo , Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA