Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 326(6): F894-F916, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38634137

RESUMEN

Mild cognitive impairment (MCI) is common in people with chronic kidney disease (CKD), and its prevalence increases with progressive loss of kidney function. MCI is characterized by a decline in cognitive performance greater than expected for an individual age and education level but with minimal impairment of instrumental activities of daily living. Deterioration can affect one or several cognitive domains (attention, memory, executive functions, language, and perceptual motor or social cognition). Given the increasing prevalence of kidney disease, more and more people with CKD will also develop MCI causing an enormous disease burden for these individuals, their relatives, and society. However, the underlying pathomechanisms are poorly understood, and current therapies mostly aim at supporting patients in their daily lives. This illustrates the urgent need to elucidate the pathogenesis and potential therapeutic targets and test novel therapies in appropriate preclinical models. Here, we will outline the necessary criteria for experimental modeling of cognitive disorders in CKD. We discuss the use of mice, rats, and zebrafish as model systems and present valuable techniques through which kidney function and cognitive impairment can be assessed in this setting. Our objective is to enable researchers to overcome hurdles and accelerate preclinical research aimed at improving the therapy of people with CKD and MCI.


Asunto(s)
Disfunción Cognitiva , Modelos Animales de Enfermedad , Insuficiencia Renal Crónica , Animales , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/psicología , Insuficiencia Renal Crónica/complicaciones , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Humanos , Ratones , Pez Cebra , Cognición , Ratas , Riñón/fisiopatología , Riñón/metabolismo
2.
Exp Physiol ; 108(3): 480-490, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36644793

RESUMEN

NEW FINDINGS: What is the central question of this study? Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce cardiovascular risk in patients with both diabetic and non-diabetic kidney disease: can SGLT2 inhibition improve renal pressure natriuresis (PN), an important mechanism for long-term blood pressure control, which is impaired in type 1 diabetes mellitus (T1DM)? What is the main finding and its importance? The SGLT2 inhibitor dapagliflozin did not enhance the acute in vivo PN response in either healthy or T1DM Sprague-Dawley rats. The data suggest that the mechanism underpinning the clinical benefits of SGLT2 inhibitors on health is unlikely to be due to an enhanced natriuretic response to increased blood pressure. ABSTRACT: Type 1 diabetes mellitus (T1DM) leads to serious complications including premature cardiovascular and kidney disease. Hypertension contributes importantly to these adverse outcomes. The renal pressure natriuresis (PN) response, a key regulator of blood pressure (BP), is impaired in rats with T1DM as tubular sodium reabsorption fails to down-regulate with increasing BP. We hypothesised that sodium-glucose cotransporter 2 (SGLT2) inhibitors, which reduce cardiovascular risk in kidney disease, would augment the PN response in T1DM rats. Non-diabetic or T1DM (35-50 mg/kg streptozotocin i.p.) adult male Sprague-Dawley rats were anaesthetised (thiopental 50 mg/kg i.p.) and randomised to receive either dapagliflozin (1 mg/kg i.v.) or vehicle. Baseline sodium excretion was measured and then BP was increased by sequential arterial ligations to induce the PN response. In non-diabetic animals, the natriuretic and diuretic responses to increasing BP were not augmented by dapagliflozin. Dapagliflozin induced glycosuria, but this was not influenced by BP. In T1DM rats the PN response was impaired. Dapagliflozin again increased urinary glucose excretion but did not enhance PN. Inhibition of SGLT2 does not enhance the PN response in rats, either with or without T1DM. SGLT2 makes only a minor contribution to tubular sodium reabsorption and does not contribute to the impaired PN response in T1DM.


Asunto(s)
Diabetes Mellitus Tipo 1 , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Masculino , Ratas , Glucemia , Presión Sanguínea/fisiología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Glucosa , Natriuresis , Ratas Sprague-Dawley , Sodio , Transportador 2 de Sodio-Glucosa , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
3.
Physiology (Bethesda) ; 36(1): 21-34, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33325814

RESUMEN

Blood pressure follows a daily rhythm, dipping during nocturnal sleep in humans. Attenuation of this dip (nondipping) is associated with increased risk of cardiovascular disease. Renal control of sodium homeostasis is essential for long-term blood pressure control. Sodium reabsorption and excretion have rhythms that rely on predictive/circadian as well as reactive adaptations. We explore how these rhythms might contribute to blood pressure rhythm in health and disease.


Asunto(s)
Hipertensión , Sodio , Presión Sanguínea , Ritmo Circadiano , Humanos , Riñón
4.
Kidney Int ; 102(5): 1115-1126, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35998848

RESUMEN

Cardiovascular disease is a complication of systemic inflammatory diseases including anti-neutrophil cytoplasm antibody-associated vasculitis (AAV). The mechanisms of cardiovascular morbidity in AAV are poorly understood, and risk-reduction strategies are lacking. Therefore, in a series of double-blind, randomized case-control forearm plethysmography and crossover systemic interventional studies, we examined arterial stiffness and endothelial function in patients with AAV in long-term disease remission and in matched healthy volunteers (32 each group). The primary outcome for the case-control study was the difference in endothelium-dependent vasodilation between health and AAV, and for the crossover study was the difference in pulse wave velocity (PWV) between treatment with placebo and selective endothelin-A receptor antagonism. Parallel in vitro studies of circulating monocytes and platelets explored mechanisms. Compared to healthy volunteers, patients with AAV had 30% reduced endothelium-dependent vasodilation and 50% reduced acute release of endothelial active tissue plasminogen activator (tPA), both significant in the case-control study. Patients with AAV had significantly increased arterial stiffness (PWV: 7.3 versus 6.4 m/s). Plasma endothelin-1 was two-fold higher in AAV and independently predicted PWV and tPA release. Compared to placebo, both selective endothelin-A and dual endothelin-A/B receptor blockade reduced PWV and increased tPA release in AAV in the crossover study. Mechanistically, patients with AAV had increased platelet activation, more platelet-monocyte aggregates, and altered monocyte endothelin receptor function, reflecting reduced endothelin-1 clearance. Patients with AAV in long-term remission have elevated cardiovascular risk and endothelin-1 contributes to this. Thus, our data support a role for endothelin-blockers to reduce cardiovascular risk by reducing arterial stiffness and increasing circulating tPA activity.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos , Enfermedades Cardiovasculares , Rigidez Vascular , Humanos , Activador de Tejido Plasminógeno , Fibrinólisis , Análisis de la Onda del Pulso , Enfermedades Cardiovasculares/etiología , Endotelina-1 , Estudios de Casos y Controles , Estudios Cruzados , Factores de Riesgo , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/complicaciones , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/tratamiento farmacológico , Factores de Riesgo de Enfermedad Cardiaca , Receptores de Endotelina
5.
Kidney Int ; 100(2): 272-275, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34294206

RESUMEN

Salbutamol activates the NaCl cotransporter of the distal convoluted tubule. Salbutamol, in conjunction with high salt intake, induced hypertension in mice, rescued by thiazide therapy. Phosphoproteomics identified protein phosphatase 1/inhibitor 1 as a distinct regulatory node for NaCl cotransporter activation by salbutamol, which did not activate the transporter in inhibitor 1 knockout mice. Salbutamol is widely used in respiratory medicine, and the acquisition of salt sensitivity may be relevant to understanding cardiovascular risk in certain patients.


Asunto(s)
Hipertensión , Cloruro de Sodio Dietético , Albuterol/efectos adversos , Animales , Humanos , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Túbulos Renales Distales , Ratones , Cloruro de Sodio , Miembro 3 de la Familia de Transportadores de Soluto 12
6.
Foodborne Pathog Dis ; 18(11): 784-789, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34287066

RESUMEN

With over 1 million estimated cases per year in the United States, foodborne salmonellosis is an important public health issue. Chicken products are frequent sources of foodborne Salmonella infection. These bacteria readily colonize the gastrointestinal tract of broiler chickens, and feed is a known vector. Past research has demonstrated that the survivability of Salmonella in feed is dependent on the serovar and strain. Therefore, the objective of this research was to compare colonization incidence of these two serovars in broiler chicken tissues by administration of feed contaminated with Salmonella enterica serovar Enteritidis (SE) or Salmonella enterica serovar Heidelberg (SH). A comparison was made with equal conditions so that there was no influence of other factors. Birds were inoculated by addition of Salmonella to the feed (1 × 104 colony-forming unit [CFU]/g of feed) at 14 days of age, and the following tissue samples were collected from each bird after grow-out (days 34-41 depending on the trial): abdominal cavity swab, bone marrow swab, cloaca swab, lung swab, breast, bursa and thymus, ceca, crop, kidney, liver and spleen, skin, spinal cord, thigh, and trachea. A higher percentage of birds inoculated with SE were positive in at least one tissue compared with SH (68% and 9%, respectively), and the SE inoculated birds also showed a higher number of positive tissue samples than SH (13.1% and 0.7%, respectively). Recovery of SH was low for all tissue samples. However, recovery of SE was variable between samples, with ceca showing the highest percentage (50%). These results indicate that challenge at day 14 through feed administration results in greater colonization by SE compared with SH, suggesting that monitoring and control methods for Salmonella in feed should focus on SE to have the greatest positive effect.


Asunto(s)
Enfermedades de las Aves de Corral , Salmonelosis Animal , Animales , Ciego , Pollos , Salmonella enteritidis , Serogrupo
7.
Kidney Int ; 98(5): 1193-1209, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32569653

RESUMEN

The endothelin system may be an important player in hypertensive end-organ injury as endothelin-1 increases blood pressure and is pro-inflammatory. The immune system is emerging as an important regulator of blood pressure and we have shown that the early hypertensive response to angiotensin-II infusion was amplified in mice deficient of myeloid endothelin-B (ETB) receptors (LysM-CreEdnrblox/lox). Hypothesizing that these mice would display enhanced organ injury, we gave angiotensin-II to LysM-CreEdnrblox/lox and littermate controls (Ednrblox/lox) for six weeks. Unexpectedly, LysM-CreEdnrblox/lox mice were significantly protected from organ injury, with less proteinuria, glomerulosclerosis and inflammation of the kidney compared to controls. In the eye, LysM-CreEdnrblox/lox mice had fewer retinal hemorrhages, less microglial activation and less vessel rarefaction. Cardiac remodeling and dysfunction were similar in both groups at week six but LysM-CreEdnrblox/lox mice had better endothelial function. Although blood pressure was initially higher in LysM-CreEdnrblox/lox mice, this was not sustained. A natriuretic switch at about two weeks, due to enhanced ETB signaling in the kidney, induced a hypertensive reversal. By week six, blood pressure was lower in LysM-CreEdnrblox/lox mice than in controls. At six weeks, macrophages from LysM-CreEdnrblox/lox mice were more anti-inflammatory and had greater phagocytic ability compared to the macrophages of Ednrblox/lox mice. Thus, myeloid cell ETB receptor signaling drives this injury both through amplifying hypertension and by inflammatory polarization of macrophages.


Asunto(s)
Angiotensina II , Hipertensión , Animales , Presión Sanguínea , Endotelinas , Hipertensión/inducido químicamente , Hipertensión/genética , Riñón , Ratones , Receptor de Endotelina B/genética
8.
Eur Heart J ; 40(9): 768-784, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30657897

RESUMEN

AIMS: Hypertension is common. Recent data suggest that macrophages (Mφ) contribute to, and protect from, hypertension. Endothelin-1 (ET-1) is the most potent endogenous vasoconstrictor with additional pro-inflammatory properties. We investigated the role of the ET system in experimental and clinical hypertension by modifying Mφ number and phenotype. METHODS AND RESULTS: In vitro, Mφ ET receptor function was explored using pharmacological, gene silencing, and knockout approaches. Using the CD11b-DTR mouse and novel mice with myeloid cell-specific endothelin-B (ETB) receptor deficiency (LysMETB-/-), we explored the effects of modifying Mφ number and phenotype on the hypertensive effects of ET-1, angiotensin II (ANG II), a model that is ET-1 dependent, and salt. In patients with small vessel vasculitis, the impacts of Mφ depleting and non-depleting therapies on blood pressure (BP) and endothelial function were examined. Mouse and human Mφ expressed both endothelin-A and ETB receptors and displayed chemokinesis to ET-1. However, stimulation of Mφ with exogenous ET-1 did not polarize Mφ phenotype. Interestingly, both mouse and human Mφ cleared ET-1 through ETB receptor mediated, and dynamin-dependent, endocytosis. Mφ depletion resulted in an augmented chronic hypertensive response to both ET-1 and salt. LysMETB-/- mice displayed an exaggerated hypertensive response to both ET-1 and ANG II. Finally, in patients who received Mφ depleting immunotherapy BP was higher and endothelial function worse than in those receiving non-depleting therapies. CONCLUSION: Mφ and ET-1 may play an important role in BP control and potentially have a critical role as a therapeutic target in hypertension.


Asunto(s)
Angiotensina II/fisiología , Endotelina-1/fisiología , Hipertensión/patología , Hipertensión/fisiopatología , Macrófagos/fisiología , Receptor de Endotelina B/fisiología , Animales , Modelos Animales de Enfermedad , Endocitosis/fisiología , Humanos , Hipertensión/etiología , Ratones , Receptor de Endotelina A
9.
J Physiol ; 597(3): 767-780, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30537108

RESUMEN

KEY POINTS: Type 1 diabetes mellitus increases cardiovascular risk; hypertension amplifies this risk, while pressure natriuresis regulates long-term blood pressure. We induced type 1 diabetes in rats by streptozotocin injection and demonstrated a substantial impairment of pressure natriuresis: acute increases in blood pressure did not increase renal medullary blood flow, tubular sodium reabsorption was not downregulated, and proximal tubule sodium reabsorption, measured by lithium clearance, was unaffected. Insulin reduced blood glucose in diabetic rats, and rescued the pressure natriuresis response without influencing lithium clearance, but did not restore medullary blood flow. Radiotelemetry showed that diastolic blood pressure was increased in diabetic rats, and its diurnal variation was reduced. Increases in medullary blood flow and decreases in distal tubule sodium reabsorption that offset acute rises in BP are impaired in early type 1 diabetes, and this impairment could be a target for preventing hypertension in type 1 diabetes. ABSTRACT: Type 1 diabetes mellitus (T1DM) substantially increases cardiovascular risk, and hypertension amplifies this risk. Blood pressure (BP) and body sodium homeostasis are linked. T1DM patients have increased total exchangeable sodium, correlating directly with BP. Pressure natriuresis is an important physiological regulator of BP. We hypothesised that pressure natriuresis would be impaired, and BP increased, in the early phase of T1DM. Male Sprague-Dawley rats were injected with streptozotocin (30-45 mg/kg) or citrate vehicle. After 3 weeks, pressure natriuresis was induced by serial arterial ligation. In non-diabetic controls, this increased fractional excretion of sodium from ∼1% to ∼25% of the filtered load (P < 0.01); in T1DM rats, the response was significantly blunted, peaking at only ∼3% (P < 0.01). Mechanistically, normal lithium clearance suggested that distal tubule sodium reabsorption was not downregulated with increased BP in T1DM rats. The pressure dependence of renal medullary perfusion, considered a key factor in the integrated response, was abolished. Insulin therapy rescued the natriuretic response in diabetic rats, restoring normal downregulation of tubular sodium reabsorption when BP was increased. However, the pressure dependence of medullary perfusion was not restored, suggesting persistent vascular dysfunction despite glycaemic control. Radiotelemetry showed that T1DM did not affect systolic BP, but mean diastolic BP was ∼5 mmHg higher than in non-diabetic controls (P < 0.01), and normal diurnal variation was reduced. In conclusion, functional impairment of renal sodium and BP homeostasis is an early manifestation of T1DM, preceding hypertension and nephropathy. Early intervention to restore pressure natriuresis in T1DM may complement reductions in cardiovascular risk achieved with glycaemic control.


Asunto(s)
Presión Sanguínea/fisiología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/fisiopatología , Natriuresis/fisiología , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Regulación hacia Abajo/fisiología , Hemodinámica/fisiología , Hipertensión/fisiopatología , Riñón/metabolismo , Riñón/fisiopatología , Litio/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Circulación Renal/fisiología , Sodio/metabolismo
10.
Am J Physiol Renal Physiol ; 317(6): F1536-F1548, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31588796

RESUMEN

The sodium-chloride cotransporter (NCC) in the distal convoluted tubule contributes importantly to sodium balance and blood pressure (BP) regulation. NCC phosphorylation determines transport activity and has a diurnal rhythm influenced by glucocorticoids. Disturbing this rhythm induces "nondipping" BP, an abnormality that increases cardiovascular risk. The receptor through which glucocorticoids regulate NCC is not known. In this study, we found that acute administration of corticosterone to male C57BL6 mice doubled NCC phosphorylation without affecting total NCC abundance in both adrenalectomized and adrenal-intact mice. Corticosterone also increased the whole kidney expression of canonical clock genes: period circadian protein homolog 1 (Per1), Per2, cryptochrome 1, and aryl hydrocarbon receptor nuclear translocator-like protein 1. In adrenal-intact mice, chronic blockade of glucocorticoid receptor (GR) with RU486 did not change total NCC but prevented corticosterone-induced NCC phosphorylation and activation of clock genes. Blockade of mineralocorticoid receptor (MR) with spironolactone reduced the total pool of NCC but did not affect stimulation by corticosterone. The diurnal rhythm of NCC phosphorylation, measured at 6-h intervals, was blunted by chronic GR blockade, and a similar dampening of diurnal variation was seen in GR heterozygous null mice. These effects on NCC phosphorylation did not reflect altered rhythmicity of plasma corticosterone or serum and glucocorticoid-induced kinase 1 activity. Both mineralocorticoids and glucocorticoids emerge as regulators of NCC, acting via distinct receptor pathways. MR activation provides maintenance of the NCC protein pool; GR activation dynamically regulates NCC phosphorylation and establishes the diurnal rhythm of NCC activity. This study has implications for circadian BP homeostasis, particularly in individuals with abnormal glucocorticoid signaling as is found in chronic stress and corticosteroid therapy.


Asunto(s)
Ritmo Circadiano/fisiología , Riñón/metabolismo , Receptores de Glucocorticoides/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Adrenalectomía , Animales , Proteínas CLOCK/biosíntesis , Proteínas CLOCK/genética , Ritmo Circadiano/efectos de los fármacos , Corticosterona/sangre , Corticosterona/farmacología , Riñón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Mifepristona/farmacología , Antagonistas de Receptores de Mineralocorticoides/farmacología , Fosforilación/efectos de los fármacos , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/efectos de los fármacos , Espironolactona/farmacología
11.
Nephrol Dial Transplant ; 34(Suppl 3): iii2-iii11, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31800080

RESUMEN

There have been significant recent advances in our understanding of the mechanisms that maintain potassium homoeostasis and the clinical consequences of hyperkalemia. In this article we discuss these advances within a concise review of the pathophysiology, risk factors and consequences of hyperkalemia. We highlight aspects that are of particular relevance for clinical practice. Hyperkalemia occurs when renal potassium excretion is limited by reductions in glomerular filtration rate, tubular flow, distal sodium delivery or the expression of aldosterone-sensitive ion transporters in the distal nephron. Accordingly, the major risk factors for hyperkalemia are renal failure, diabetes mellitus, adrenal disease and the use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers or potassium-sparing diuretics. Hyperkalemia is associated with an increased risk of death, and this is only in part explicable by hyperkalemia-induced cardiac arrhythmia. In addition to its well-established effects on cardiac excitability, hyperkalemia could also contribute to peripheral neuropathy and cause renal tubular acidosis. Hyperkalemia-or the fear of hyperkalemia-contributes to the underprescription of potentially beneficial medications, particularly in heart failure. The newer potassium binders could play a role in attempts to minimize reduced prescribing of renin-angiotensin inhibitors and mineraolocorticoid antagonists in this context.


Asunto(s)
Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/complicaciones , Hiperpotasemia , Potasio/sangre , Insuficiencia Renal/complicaciones , Salud Global , Tasa de Filtración Glomerular , Homeostasis , Humanos , Hiperpotasemia/sangre , Hiperpotasemia/epidemiología , Hiperpotasemia/etiología , Incidencia , Insuficiencia Renal/sangre , Insuficiencia Renal/fisiopatología , Factores de Riesgo
12.
Exp Physiol ; 108(2): 163-164, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36537861
13.
Circulation ; 133(14): 1360-70, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26951843

RESUMEN

BACKGROUND: The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11ßHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11ßHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. METHODS AND RESULTS: Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. CONCLUSIONS: Reduced 11ßHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11ßHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor-dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Ansia/fisiología , Hipertensión/genética , Síndrome de Exceso Aparente de Mineralocorticoides/fisiopatología , Proteínas del Tejido Nervioso/deficiencia , Receptores de Mineralocorticoides/fisiología , Cloruro de Sodio Dietético/toxicidad , Núcleo Solitario/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/fisiología , Animales , Barorreflejo/efectos de los fármacos , Corticosterona/sangre , Dexametasona/farmacología , Conducta de Ingestión de Líquido , Genes Sintéticos , Hipertensión/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Exceso Aparente de Mineralocorticoides/tratamiento farmacológico , Síndrome de Exceso Aparente de Mineralocorticoides/genética , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Nefronas/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Nestina/genética , Neuronas/fisiología , Potasio/orina , ARN Mensajero/biosíntesis , Reflejo Anormal , Núcleo Solitario/fisiopatología , Espironolactona/farmacología
14.
Kidney Int ; 91(2): 315-323, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27780585

RESUMEN

Nucleotides are key subunits for nucleic acids and provide energy for intracellular metabolism. They can also be released from cells to act physiologically as extracellular messengers or pathologically as danger signals. Extracellular nucleotides stimulate membrane receptors in the P2 and P1 family. P2X are ATP-activated cation channels; P2Y and P1 are G-protein coupled receptors activated by ATP, ADP, UTP, and UDP in the case of P2 or adenosine for P1. Renal P2 receptors influence both vascular contractility and tubular function. Renal cells also express ectonucleotidases that rapidly hydrolyze extracellular nucleotides. These enzymes integrate this multireceptor purinergic-signaling complex by determining the nucleotide milieu to titrate receptor activation. Purinergic signaling also regulates immune cell function by modulating the synthesis and release of various cytokines such as IL1-ß and IL-18 as part of inflammasome activation. Abnormal or excessive stimulation of this intricate paracrine system can be pro- or anti-inflammatory, and is also linked to necrosis and apoptosis. Kidney tissue injury causes a localized increase in ATP concentration, and sustained activation of P2 receptors can lead to renal glomerular, tubular, and vascular cell damage. Purinergic receptors also regulate the activity and proliferation of fibroblasts, promoting both inflammation and fibrosis in chronic disease. In this short review we summarize some of the recent findings related to purinergic signaling in the kidney. We focus predominantly on the P2X7 receptor, discussing why antagonists have so far disappointed in clinical trials and how advances in our understanding of purinergic signaling might help to reposition these compounds as potential treatments for renal disease.


Asunto(s)
Adenosina/metabolismo , Enfermedades Renales/metabolismo , Riñón/metabolismo , Nucleótidos de Purina/metabolismo , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Transducción de Señal , Animales , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Ligandos , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Receptores Purinérgicos P1/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2X7/efectos de los fármacos , Receptores Purinérgicos P2X7/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Curr Hypertens Rep ; 19(12): 100, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29138984

RESUMEN

The metabolic syndrome describes a clustering of risk factors-visceral obesity, dyslipidaemia, insulin resistance, and salt-sensitive hypertension-that increases mortality related to cardiovascular disease, type 2 diabetes, cancer, and non-alcoholic fatty liver disease. The prevalence of these concurrent comorbidities is ~ 25-30% worldwide, and metabolic syndrome therefore presents a significant global public health burden. Evidence from clinical and preclinical studies indicates that glucocorticoid excess is a key causal feature of metabolic syndrome. This is not increased systemic in circulating cortisol, rather increased bioavailability of active glucocorticoids within tissues. This review examines the role of covert glucocorticoid excess on the hypertension of the metabolic syndrome. Here, the role of the 11ß-hydroxysteroid dehydrogenase enzymes, which exert intracrine and paracrine control over glucocorticoid signalling, is examined. 11ßHSD1 amplifies glucocorticoid action in cells and contributes to hypertension through direct and indirect effects on the kidney and vasculature. The deactivation of glucocorticoid by 11ßHSD2 controls ligand access to glucocorticoid and mineralocorticoid receptors: loss of function promotes salt retention and hypertension. As for hypertension in general, high blood pressure in the metabolic syndrome reflects a complex interaction between multiple systems. The clear association between high dietary salt, glucocorticoid production, and metabolic disorders has major relevance for human health and warrants systematic evaluation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasas/fisiología , Hipertensión/fisiopatología , Síndrome Metabólico/fisiopatología , Animales , Glucocorticoides/fisiología , Humanos , Receptores de Glucocorticoides/fisiología , Receptores de Mineralocorticoides/fisiología , Sodio/metabolismo
16.
J Am Soc Nephrol ; 27(11): 3345-3355, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27020854

RESUMEN

Extracellular vesicles (ECVs) facilitate intercellular communication along the nephron, with the potential to change the function of the recipient cell. However, it is not known whether this is a regulated process analogous to other signaling systems. We investigated the potential hormonal regulation of ECV transfer and report that desmopressin, a vasopressin analogue, stimulated the uptake of fluorescently loaded ECVs into a kidney collecting duct cell line (mCCDC11) and into primary cells. Exposure of mCCDC11 cells to ECVs isolated from cells overexpressing microRNA-503 led to downregulated expression of microRNA-503 target genes, but only in the presence of desmopressin. Mechanistically, ECV entry into mCCDC11 cells required cAMP production, was reduced by inhibiting dynamin, and was selective for ECVs from kidney tubular cells. In vivo, we measured the urinary excretion and tissue uptake of fluorescently loaded ECVs delivered systemically to mice before and after administration of the vasopressin V2 receptor antagonist tolvaptan. In control-treated mice, we recovered 2.5% of administered ECVs in the urine; tolvaptan increased recovery five-fold and reduced ECV deposition in kidney tissue. Furthermore, in a patient with central diabetes insipidus, desmopressin reduced the excretion of ECVs derived from glomerular and proximal tubular cells. These data are consistent with vasopressin-regulated uptake of ECVs in vivo We conclude that ECV uptake is a specific and regulated process. Physiologically, ECVs are a new mechanism of intercellular communication; therapeutically, ECVs may be a vehicle by which RNA therapy could be targeted to specific cells for the treatment of kidney disease.


Asunto(s)
Vesículas Extracelulares/fisiología , Túbulos Renales Colectores/citología , Vasopresinas/fisiología , Adolescente , Animales , Desamino Arginina Vasopresina/farmacología , Vesículas Extracelulares/efectos de los fármacos , Humanos , Túbulos Renales Colectores/ultraestructura , Masculino , Ratones , Ratas
17.
J Am Soc Nephrol ; 26(7): 1537-48, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25349206

RESUMEN

Na(+) transport in the renal distal convoluted tubule (DCT) by the thiazide-sensitive NaCl cotransporter (NCC) is a major determinant of total body Na(+) and BP. NCC-mediated transport is stimulated by aldosterone, the dominant regulator of chronic Na(+) homeostasis, but the mechanism is controversial. Transport may also be affected by epithelial remodeling, which occurs in the DCT in response to chronic perturbations in electrolyte homeostasis. Hsd11b2(-/-) mice, which lack the enzyme 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2) and thus exhibit the syndrome of apparent mineralocorticoid excess, provided an ideal model in which to investigate the potential for DCT hypertrophy to contribute to Na(+) retention in a hypertensive condition. The DCTs of Hsd11b2(-/-) mice exhibited hypertrophy and hyperplasia and the kidneys expressed higher levels of total and phosphorylated NCC compared with those of wild-type mice. However, the striking structural and molecular phenotypes were not associated with an increase in the natriuretic effect of thiazide. In wild-type mice, Hsd11b2 mRNA was detected in some tubule segments expressing Slc12a3, but 11ßHSD2 and NCC did not colocalize at the protein level. Thus, the phosphorylation status of NCC may not necessarily equate to its activity in vivo, and the structural remodeling of the DCT in the knockout mouse may not be a direct consequence of aberrant corticosteroid signaling in DCT cells. These observations suggest that the conventional concept of mineralocorticoid signaling in the DCT should be revised to recognize the complexity of NCC regulation by corticosteroids.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/farmacología , Túbulos Renales Distales/patología , Fosforilación/efectos de los fármacos , Simportadores del Cloruro de Sodio/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , ADN Complementario/análisis , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Hipertrofia/patología , Túbulos Renales Distales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa/métodos , ARN/análisis , Distribución Aleatoria , Transcitosis/fisiología
18.
Kidney Int ; 88(5): 1079-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26108066

RESUMEN

Chronic activation of the renin-angiotensin system promotes hypertension, renal microvascular dysfunction, tissue hypoxia, and inflammation. Despite similar hypertension, an injurious response to excess angiotensin II is greater in F344 than in Lewis rats; the latter displaying renoprotection. Here we studied whether p2rx7, encoding the P2X7 receptor (P2X7R), is a candidate gene for the differential susceptibility to vascular dysfunction under high angiotensin II tone. A 14-day infusion of angiotensin II into F344 rats increased blood pressure by about 15 mm Hg without inducing fibrosis or albuminuria. In vivo pressure natriuresis was suppressed, medullary perfusion reduced by half, and the corticomedullary oxygenation gradient disrupted. Selective P2X7R antagonism restored pressure natriuresis, promoting a significant leftward shift in the intercept and increasing the slope. Sodium excretion was increased sixfold and blood pressure normalized. The specific P2X7R antagonist AZ11657312 increased renal medullary perfusion, but only in angiotensin II-treated rats. Tissue oxygenation was improved by P2X7R blockade, particularly in poorly oxygenated regions of the kidney. Thus, activation of P2X7R induces microvascular dysfunction and regional hypoxia when angiotensin II is elevated and these effects may contribute to progression of renal injury induced by chronic angiotensin II.


Asunto(s)
Corteza Renal/irrigación sanguínea , Médula Renal/irrigación sanguínea , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/metabolismo , Circulación Renal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Endotelio Vascular/metabolismo , Expresión Génica/efectos de los fármacos , Corteza Renal/fisiología , Médula Renal/fisiología , Masculino , Natriuresis/efectos de los fármacos , Óxido Nítrico/metabolismo , Oxígeno/sangre , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores Purinérgicos P2X7/genética , Vasoconstrictores/farmacología
20.
Exp Physiol ; 100(11): 1362-78, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26337786

RESUMEN

NEW FINDINGS: What is the central question of this study? Glucocorticoids act in the kidney to promote salt and water retention. Renal 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1), by increasing local concentrations of glucocorticoids, may exert an antinatriuretic effect. We hypothesized that global deletion of 11ßHSD1 in the mouse would give rise to a salt-wasting renal phenotype. What is the main finding and its importance? We subjected a mouse model of global 11ßHSD1 deletion to studies of water and electrolyte balance, renal clearance, urinary steroid excretion, renin-angiotensin system activation and renal sodium transporter expression. We found no significant effects on renal sodium or water excretion. Any effect of renal 11ßHSD1 on sodium homeostasis is subtle. Glucocorticoids act in the kidney to regulate glomerular haemodynamics and tubular sodium transport; the net effect favours sodium retention. 11ß-Hydroxysteroid dehydrogenase type 1 (11ßHSD1) is expressed in the renal tubules and the interstitial cells of the medulla, where it is likely to regenerate active glucocorticoids from inert 11-keto forms. The physiological function of renal 11ßHSD1 is largely unknown. We hypothesized that loss of renal 11ßHSD1 would result in salt wasting and tested this in a knockout mouse model in which 11ßHSD1 was deleted in all body tissues. In balance studies, 11ßHSD1 deletion had no effect on water, sodium or potassium metabolism; transition to a low-sodium diet did not reveal a natriuretic phenotype. Renal clearance studies demonstrated identical haemodynamic parameters (arterial blood pressure, renal blood flow and glomerular filtration rate) in knockout and wild-type mice, but revealed an augmented kaliuretic response to thiazides in 11ßHSD1 knockout animals. There was no effect on the natriuretic response to the amiloride analogue benzamil. Urinary excretion of deoxycorticosterone was higher in 11ßHSD1 knockout mice, and there was hypertrophy of cells in the zona fasciculata of the adrenal cortex. There was no difference in the activity of the renin-angiotensin and nitric oxide systems, no difference in renal histology and no difference in the abundance of key tubular transporter proteins. We conclude that any effect of 11ßHSD1 on renal sodium excretion is subtle.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Homeostasis , Riñón/fisiología , Sodio/fisiología , Animales , Glucocorticoides/fisiología , Ratones Noqueados , Potasio/metabolismo , Sistema Renina-Angiotensina , Sodio/metabolismo , Equilibrio Hidroelectrolítico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA