Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(9)2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37175599

RESUMEN

The Na+-activated Na+ channel (Nax) and salt-inducible kinase (SIK) are stimulated by increases in local Na+ concentration, affecting (Na+ + K+)-ATPase activity. To test the hypothesis that the triad Nax/SIK/(Na+ + K+)-ATPase contributes to kidney injury and salt-sensitive hypertension (HTN), uninephrectomized male Wistar rats (200 g; n = 20) were randomly divided into 4 groups based on a salt diet (normal salt diet; NSD-0.5% NaCl-or high-salt diet; HSD-4% NaCl) and subcutaneous administration of saline (0.9% NaCl) or deoxycorticosterone acetate (DOCA, 8 mg/kg), as follows: Control (CTRL), CTRL-Salt, DOCA, and DOCA-Salt, respectively. After 28 days, the following were measured: kidney function, blood pressure, (Na+ + K+)-ATPase and SIK1 kidney activities, and Nax and SIK1 renal expression levels. SIK isoforms in kidneys of CTRL rats were present in the glomerulus and tubular epithelia; they were not altered by HSD and/or HTN. CTRL-Salt rats remained normotensive but presented slight kidney function decay. HSD rats displayed augmentation of the Nax/SIK/(Na+ + K+)-ATPase pathway. HTN, kidney injury, and kidney function decay were present in all DOCA rats; these were aggravated by HSD. DOCA rats presented unaltered (Na+ + K+)-ATPase activity, diminished total SIK activity, and augmented SIK1 and Nax content in the kidney cortex. DOCA-Salt rats expressed SIK1 activity and downregulation in (Na+ + K+)-ATPase activity in the kidney cortex despite augmented Nax content. The data of this study indicate that the (Na+ + K+)-ATPase activity response to SIK is attenuated in rats under HSD, independent of HTN, as a mechanism contributing to kidney injury and salt-sensitive HTN.


Asunto(s)
Acetato de Desoxicorticosterona , Hipertensión , Ratas , Masculino , Animales , Cloruro de Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Ratas Wistar , Hipertensión/metabolismo , Sodio/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo , Presión Sanguínea , Riñón/metabolismo , Iones/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
2.
Curr Hypertens Rev ; 18(2): 91-100, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35170417

RESUMEN

The production of renin by the principal cells of the collecting duct has widened our understanding of the regulation of intrarenal angiotensin II (Ang II) generation and blood pressure. In the collecting duct, Ang II increases the synthesis and secretion of renin by mechanisms involving the activation of Ang II type 1 receptor (AT1R) via stimulation of the PKCα, Ca2+, and cAMP/PKA/CREB pathways. Additionally, paracrine mediators, including vasopressin (AVP), prostaglandins, bradykinin (BK), and atrial natriuretic peptide (ANP), regulate renin in principal cells. During Ang II-dependent hypertension, despite plasma renin activity suppression, renin and prorenin receptor (RPR) are upregulated in the collecting duct and promote de novo formation of intratubular Ang II. Furthermore, activation of PRR by its natural agonists, prorenin and renin, may contribute to the stimulation of profibrotic factors independent of Ang II. Thus, the interactions of RAS components with paracrine hormones within the collecting duct enable tubular compartmentalization of the RAS to orchestrate complex mechanisms that increase intrarenal Ang II, Na+ reabsorption, and blood pressure.


Asunto(s)
Hipertensión , Renina , Humanos , Angiotensina II , Presión Sanguínea , Receptor de Prorenina , Receptores de Superficie Celular/metabolismo , Sistema Renina-Angiotensina
3.
Nutr Neurosci ; 25(9): 1973-1994, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33871318

RESUMEN

Undernutrition is characterized by an imbalance of essential nutrients with an insufficient nutritional intake, a disorder in which the clinical manifestations in most cases are the result of the economic and social context in which the individual lives. In 1990, the study by the medical and humanitarian Naíde Teodósio (1915-2005) and coworkers, which formulated the Regional Basic Diet (RBD) model for inducing undernutrition, was published. This diet model took its origin from the observation of the dietary habits of families that inhabited impoverished areas from the Pernambuco State. RBD mimics an undernutrition framework that extends not only to the Brazilian population, but to populations in different regions worldwide. The studies based on RBD-induced deficiencies provide a better understanding of the impact of undernutrition on the pathophysiological mechanisms underlying the most diverse prevalent diseases. Indexed papers that are analyzed in this review focus on the importance of using RBD in different areas of knowledge. These papers reflect a new paradigm in translational medicine: they show how the study of pathology using the RBD model in animals over the past 30 years has and still can help scientists today, shedding light on the mechanisms of prevalent diseases that affect impoverished populations.


Asunto(s)
Desnutrición , Animales , Brasil , Dieta , Conducta Alimentaria , Desnutrición/epidemiología
4.
Toxicon ; 199: 117-126, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34116084

RESUMEN

There is no consensus on whether serotherapy prevents acute kidney injury (AKI) and there is no pharmacotherapy to impede the disease. We aimed to elaborate an AKI model induced by the administration of Bothrops jararacussu (Bj) venom for preclinical studies. Male Wistar rats were randomly divided into 3 different groups: (1) Bj-IV: intravenous administration of 0.4 mg/kg Bj; (2) Bj-IP: intraperitoneal administration of 2.0 mg/kg Bj; (3) Bj-IM: intramuscular administration of 3.5 mg/kg Bj. For each corresponding control group, a 0.9% saline solution was administered. Kidneys, blood and urine samples were collected 24 or 72 h after administration of the Bj venom for renal function analysis. The IV- and IP-Bj groups presented a moderate tubular injury (score 3) and a time-dependent kidney dysfunction. In the Bj-IM group, renal tubular injury was aggravated (score 4) with collagen deposition and renal dysfunction was observed in the first 24 h: hyperfiltration, proteinuria, albuminuria and decreased fractional sodium excretion (FENa), regardless of the administered dose. Over time, the glomerular lesion was intensified, with a decrease in glomerular filtration rate (GFR; 67%), blood urea-nitrogen (BUN; 68%) and urine volume decrease (71%). Proteinuria and tubular function returned to control levels after 72 h. We attributed the pronounced kidney injury and reduced filtration function in the Bj-IM to the muscle damage provoked by the IM administration. We concluded that the Bj-IM is the best preclinical model of AKI with the monitoring of the progression of renal function in the periods of 24 and 72 h.


Asunto(s)
Lesión Renal Aguda , Bothrops , Venenos de Crotálidos , Lesión Renal Aguda/inducido químicamente , Animales , Venenos de Crotálidos/toxicidad , Tasa de Filtración Glomerular , Riñón , Masculino , Ratas , Ratas Wistar
5.
Toxicon ; 199: 20-30, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34058237

RESUMEN

Acute kidney injury is one of the main complications of ophidian accidents and the leading cause of death in patients who survive the initial damage effects of venom. The hypothesis proposed in this investigation is that the pharmacological repositioning of doxycycline (doxy) attenuates renal injury provoked by Bothrops jararacussu (Bj) venom. Male Wistar rats were subjected or not (control) to experimental envenomation with Bj venom (3.5 mg/kg, im). Doxy (3 mg/kg, ip) was administered 2 h after envenoming. Envenomation with Bj venom promoted tissue damage in the renal cortex (moderate degree, score 3) in 24 h associated with decreased glomerular and tubular function, which promoted proteinuria and polyuria. Doxy treatment prevented the increase in urinary volume in 3 times, the increase in plasma creatinine in 33%, the increase in blood urea-nitrogen accumulation in 65%, the increase in urinary Na+ excretion in 2 times, marked proteinuria and kidney cortex injury induced by Bj envenomation. Bj venom promoted increase in protein content (66%) and reduction of 45% (Na++K+)-ATPase activity in the renal cortex. The enzyme was detected mainly in the luminal membrane. Doxy treatment was effective in preventing the mentioned alterations, maintaining (Na++K+)-ATPase in the basolateral membranes.


Asunto(s)
Bothrops , Venenos de Crotálidos , Animales , Venenos de Crotálidos/toxicidad , Doxiciclina , Humanos , Riñón/fisiología , Masculino , Ratas , Ratas Wistar
6.
Front Physiol ; 11: 559341, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33281610

RESUMEN

In the kidney, the stimulation of renin production by the collecting duct (CD-renin) contributes to the development of hypertension. The CD is a major nephron segment for the synthesis of nitric oxide (NO), and low NO bioavailability in the renal medulla is associated with hypertension. However, it is unknown whether NO regulates renin production in the CD. To test the hypothesis that low intrarenal NO levels stimulate the production of CD-renin, we first examined renin expression in the distal nephron segments of CD-eNOS deficient mice. In these mice, specific CD-renin immunoreactivity was increased compared to wild-type littermates; however, juxtaglomerular (JG) renin was not altered. To further assess the intracellular mechanisms involved, we then treated M-1 cells with either 1 mM L-NAME (L-arginine analog), an inhibitor of NO synthase activity, or 1 mM NONOate, a NO donor. Both treatments increased intracellular renin protein levels in M-1 cells. However, only the inhibition of NOS with L-NAME stimulated renin synthesis and secretion as reflected by the increase in Ren1C transcript and renin protein levels in the extracellular media, respectively. In addition, NONOate induced a fast mobilization of cGMP and intracellular renin accumulation. These response was partially prevented by guanylyl cyclase inhibition with ODQ (1H-[1,2,4] oxadiazolo[4,3-a]quinoxalin-1]. Accumulation of intracellular renin was blocked by protein kinase G (PKG) and protein kinase C (PKC) inhibitors. Our data indicate that low NO bioavailability increases CD-renin synthesis and secretion, which may contribute to the activation of intrarenal renin angiotensin system.

7.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 102-114, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28987762

RESUMEN

Renal ischemia-reperfusion injury (IRI) is a major cause of acute renal failure. Doxycycline (Dc) belongs to the tetracycline-class of antibiotics with demonstrated beneficial molecular effects in the brain and heart, mainly through matrix metalloproteinases inhibition (MMP). However, Dc protection of renal function has not been demonstrated. We determined whether low doses of Dc would prevent decreases in glomerular filtration rate (GFR) and maintain tubular Na+ handling in Wistar rats subjected to kidney I/R. Male Wistar rats underwent bilateral kidney ischemia for 30min followed by 24h reperfusion (I/R). Doxycycline (1, 3, and 10mg/kg, i.p.) was administered 2h before surgery. Untreated I/R rats showed a 250% increase in urine volume and proteinuria, a 60% reduction in GFR, accumulation of urea-nitrogen in the blood, and a 60% decrease in the fractional Na+ excretion due to unbalanced Na+ transporter activity. Treatment with Dc 3mg/kg maintained control levels of urine volume, proteinuria, GFR, blood urea-nitrogen, fractional Na+ excretion, and equilibrated Na+ transporter activities. The Dc protection effects on renal function were associated with kidney structure preservation and prevention of TGFß and fibronectin deposition. In vitro, total MMP activity was augmented in I/R and inhibited by 25 and 50µM Dc. In vivo, I/R augmented MMP-2 and -9 protein content without changing their activities. Doxycycline treatment downregulated total MMP activity and MMP-2 and -9 protein content. Our results suggest that treatment with low dose Dc protects from IRI, thereby preserving kidney function.


Asunto(s)
Lesión Renal Aguda/patología , Citoprotección/efectos de los fármacos , Doxiciclina/farmacología , Riñón/efectos de los fármacos , Daño por Reperfusión/patología , Lesión Renal Aguda/fisiopatología , Animales , Relación Dosis-Respuesta a Droga , Tasa de Filtración Glomerular/efectos de los fármacos , Riñón/irrigación sanguínea , Riñón/patología , Riñón/fisiología , Masculino , Ratas , Ratas Wistar , Daño por Reperfusión/fisiopatología
8.
Lipids Health Dis ; 16(1): 245, 2017 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-29246161

RESUMEN

BACKGROUND: Undernutrition during childhood leads to chronic diseases in adult life including hypertension, diabetes and chronic kidney disease. Here we explore the hypothesis that physiological alterations in the bioactive lipids pattern within kidney tissue might be involved in the progression of chronic kidney disease. METHODS: Membrane fractions from kidney homogenates of undernourished rats (RBD) were submitted to lipid extraction and analysis by thin layer chromatography and cholesterol determination. RESULTS: Kidneys from RBD rats had 25% lower cholesterol content, which disturb membrane microdomains, affecting Ca2+ homeostasis and the enzymes responsible for important lipid mediators such as phosphatidylinositol-4 kinase, sphingosine kinase, diacylglicerol kinase and phospholipase A2. We observed a decrease in phosphatidylinositol(4)-phosphate (8.8 ± 0.9 vs. 3.6 ± 0.7 pmol.mg-1.mim-1), and an increase in phosphatidic acid (2.2 ± 0.8 vs. 3.8 ± 1.3 pmol.mg-1.mim-1), being these lipid mediators involved in the regulation of key renal functions. Ceramide levels are augmented in kidney tissue from RBD rats (18.7 ± 1.4 vs. 21.7 ± 1.5 fmol.mg-1.min-1) indicating an ongoing renal lesion. CONCLUSION: Results point to an imbalance in the bioactive lipid generation with further consequences to key events related to kidney function, thus contributing to the establishment of chronic kidney disease.


Asunto(s)
Colesterol/metabolismo , Hipertensión/metabolismo , Riñón/metabolismo , Desnutrición/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Insuficiencia Renal Crónica/metabolismo , 1-Fosfatidilinositol 4-Quinasa/genética , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Animales Recién Nacidos , Ceramidas/metabolismo , Diacilglicerol Quinasa/genética , Diacilglicerol Quinasa/metabolismo , Regulación de la Expresión Génica , Hipertensión/etiología , Hipertensión/genética , Hipertensión/patología , Riñón/química , Metabolismo de los Lípidos , Masculino , Desnutrición/complicaciones , Desnutrición/genética , Desnutrición/patología , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología
9.
Curr Hypertens Rep ; 19(8): 62, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28695400

RESUMEN

The presence of renin production by the principal cells of the collecting duct has opened new perspectives for the regulation of intrarenal angiotensin II (Ang II). Angiotensinogen (AGT) and angiotensin-converting enzyme (ACE) are present in the tubular fluid coming from the proximal tubule and collecting duct. All the components needed for Ang II formation are present along the nephron, and much is known about the mechanisms regulating renin in juxtaglomerular cells (JG); however, those in the collecting duct remain unclear. Ang II suppresses renin via protein kinase C (PKC) and calcium (Ca2+) in JG cells, but in the principal cells, Ang II increases renin synthesis and release through a pathophysiological mechanism that increases further intratubular Ang II de novo formation to enhance distal Na + reabsorption. Transgenic mice overexpressing renin in the collecting duct demonstrate the role of collecting duct renin in the development of hypertension. The story became even more interesting after the discovery of a specific receptor for renin and prorenin: the prorenin receptor ((P)RR), which enhances renin activity and fully activates prorenin. The interactions between (P)RR and prorenin/renin may further increase intratubular Ang II levels. In addition to Ang II, other mechanisms have been described in the regulation of renin in the collecting duct, including vasopressin (AVP), bradykinin (BK), and prostaglandins. Current active investigations are aimed at elucidating the mechanisms regulating renin in the distal nephron segments and understand its role in the pathogenesis of hypertension.


Asunto(s)
Hipertensión/metabolismo , Hipertensión/fisiopatología , Túbulos Renales Colectores/metabolismo , Renina/metabolismo , Angiotensina II/metabolismo , Animales , Humanos , Hipertensión/etiología , Túbulos Renales Colectores/fisiopatología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/fisiopatología , Túbulos Renales Proximales/fisiopatología , Renina/biosíntesis , Sistema Renina-Angiotensina/fisiología
10.
Am J Physiol Renal Physiol ; 313(2): F440-F449, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28468964

RESUMEN

ANG II has many biological effects in renal physiology, particularly in Ca2+ handling in the regulation of fluid and solute reabsorption. It involves the systemic endocrine renin-angiotensin system (RAS), but tissue and intracrine ANG II are also known. We have shown that ANG II induces heterodimerization of its AT1 and AT2 receptors (AT1R and AT2R) to stimulate sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) activity. Thus, we investigated whether ANG II-AT1R/AT2R complex is formed and internalized, and also examined the intracellular localization of this complex to determine how its effect might be exerted on renal intracrine RAS. Living cell imaging of LLC-PK1 cells, quantification of extracellular ANG II, and use of the receptor antagonists, losartan and PD123319, showed that ANG II is internalized with AT1R/AT2R heterodimers as a complex in a microtubule-dependent and clathrin-independent manner, since colchicine-but not Pitstop2-blocked this process. This result was confirmed by an increase of ß-arrestin phosphorylation after ANG II treatment, clathrin-mediated endocytosis being dependent on dephosphorylation of ß-arrestin. Internalized ANG II colocalized with an endoplasmic reticulum (ER) marker and increased levels of AT1R, AT2R, and PKCα in ER-enriched membrane fractions. This novel evidence suggests the internalization of an ANG II-AT1/AT2 complex to target ER, where it might trigger intracellular Ca2+ responses.


Asunto(s)
Angiotensina II/metabolismo , Membrana Celular/metabolismo , Endocitosis , Retículo Endoplásmico/metabolismo , Riñón/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Riñón/efectos de los fármacos , Células LLC-PK1 , Microtúbulos/metabolismo , Complejos Multiproteicos , Fosforilación , Proteína Quinasa C-alfa/metabolismo , Transporte de Proteínas , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 2/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Porcinos , beta-Arrestinas/metabolismo
11.
Physiol Rep ; 5(7)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28373410

RESUMEN

In the collecting duct (CD), the interactions of renin angiotensin system (RAS) and kallikrein-kinin system (KKS) modulate Na+ reabsorption, volume homeostasis, and blood pressure. In this study, we used a mouse kidney cortical CD cell line (M-1 cells) to test the hypothesis that in the CD, the activation of bradykinin B2 receptor (B2R) increases renin synthesis and release. Physiological concentrations of bradykinin (BK) treatment of M-1 cells increased renin mRNA and prorenin and renin protein contents in a dose-dependent manner and increased threefold renin content in the cell culture media. These effects were mediated by protein kinase C (PKC) independently of protein kinase A (PKA) because B2R antagonism with Icatibant and PKC inhibition with calphostin C, prevented these responses, but PKA inhibition with H89 did not modify the effects elicited by the B2R activation. BK-dependent stimulation of renin gene expression in CD cells also involved nitric oxide (NO) pathway because increased cGMP levels and inhibition of NO synthase with L-NAME prevented it. Complementary renin immunohistochemical studies performed in kidneys from mice with conventional B2R knockout and conditional B2R knockout in the CD, showed marked decreased renin immunoreactivity in CD, regardless of the renin presence in juxtaglomerular cells in the knockout mice. These results indicate that the activation of B2R increases renin synthesis and release by the CD cells through PKC stimulation and NO release, which support further the interactions between the RAS and KKS.


Asunto(s)
Bradiquinina/farmacología , Corteza Renal/metabolismo , Óxido Nítrico/metabolismo , Receptor de Bradiquinina B2/metabolismo , Renina/metabolismo , Animales , Bradiquinina/análogos & derivados , Antagonistas del Receptor de Bradiquinina B2/farmacología , Línea Celular , Isoquinolinas/farmacología , Corteza Renal/citología , Corteza Renal/efectos de los fármacos , Ratones , NG-Nitroarginina Metil Éster/farmacología , Naftalenos/farmacología , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología
12.
Int J Cardiol ; 238: 43-56, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28381328

RESUMEN

BACKGROUND: We investigated whether a chronic low-protein multideficient diet (BRD) from weaning turns on cardiovascular adaptive responses that could culminate in hypertension and heart failure. METHODS AND RESULTS: Systolic pressure (SP) and heart rate (HR) were determined in CTRL (normal diet) and BRD rats. Plasma albumin, plasma urea and urinary urea excretion decreased in BRD rats. In this group, echocardiography and the Langendorff technique showed: (i) increased HR and hypertension; (ii) decreased LVDP, dP/dtmax, dP/dtmin, cardiac output, ejection fraction, stroke volume and left ventricular diameter. BRD rats were less sensitive to isoproterenol (ISO) in LVDP and dP/dtmax, with unchanged dP/dtmin; Pressure-volume relationships indicated left-oriented shifts in LVDP, SP and DP, and decreased capacitance compared to CTRL. BRD rats had higher cardiac and lung indexes, accompanied by muscle atrophy and recent ventricular-infarcted areas, higher ventricular ß1-AR content, and decreased ß2-AR and α1-AR. Propranolol treatment gave similar ISO responses in both groups, disappearance of the infarcted regions and, except for ß2-AR, recovery of normal receptor expression. BRD rats had intense stimulation of plasma membrane Ca2+-ATPase (PMCA) activity, with increased Ca2+ affinity and inhibition of the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA). Ventricular phospholamban increased and Na+/Ca2+ exchanger decreased. PMCA activity correlated with an increase in its PKC-mediated phosphorylation, overlying a decrease in PKA-catalyzed phosphorylation. Propranolol normalized PKC and PKA activities with recovery of PMCA but not SERCA. CONCLUSION: BRD triggers sympathetic exacerbation and dysfunction in Ca2+ handling, accompanied by early onset of hypertension and left ventricle congestive heart failure.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Insuficiencia Cardíaca/metabolismo , Hipertensión/metabolismo , Desnutrición/metabolismo , Deficiencia de Proteína/metabolismo , Animales , Señalización del Calcio/fisiología , Enfermedad Crónica , Dieta con Restricción de Proteínas/tendencias , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Hipertensión/etiología , Hipertensión/patología , Masculino , Desnutrición/patología , Deficiencia de Proteína/patología , Proteínas Quinasas/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo , Urea/metabolismo
13.
J Renin Angiotensin Aldosterone Syst ; 18(1): 1470320316689338, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28097883

RESUMEN

Angiotensin-(3-4) (Ang-(3-4) or Val-Tyr) is the shorter angiotensin (Ang) II-derived peptide, formed through successive hydrolysis that culminates with the release of Val-Tyr as a dipeptide. It is formed both in plasma and in kidney from Ang II and Ang III, and can be considered a component of the systemic and organ-based renin-angiotensin system. It is potently antihypertensive in humans and rats, and its concerted actions on proximal tubule cells culminate in the inhibition of fluid reabsorption, hyperosmotic urinary excretion of Na+. At the renal cell signaling level, Ang-(3-4) counteracts Ang II-type 1 receptor-mediated responses by acting as an allosteric enhancer in Ang II-type 2 receptor populations that target adenosine triphosphate-dependent Ca2+ and Na+ transporters through a cyclic adenosine monophosphate-activated protein kinase pathway.


Asunto(s)
Angiotensina II/metabolismo , Sistema Renina-Angiotensina , Animales , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Péptidos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo
14.
Physiol Rep ; 3(10)2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26508737

RESUMEN

Epidemiological and animal studies have shown that placental undernutrition impairs reproduction in adult offspring, but the underlying molecular mechanisms within the male genital tract remain unknown. Due to its special physiological characteristics in transport and the modulation of the environment to which its luminal content is exposed, we hypothesized that the vas deferens would be a highly sensitive target. The goals were to investigate whether intrauterine malnutrition affects molecular mechanisms related to Ca(2+)- and oxidative stress-modulated processes and causes structural alterations in the adult rat vas deferens that could attenuate fecundity and fertility. Male adult rats malnourished in utero had increased vas deferens weight associated with thickening of the muscular coat, a decrease in the total and haploid germ cells, a marked increase in the immature cells, and a decline in the numbers of pregnant females and total offspring per male rat. The ex vivo response of vas deferens from malnourished rats demonstrated an accentuated decrease in the contractile response to phenylephrine. The vas deferens had a marked decrease in Ca(2+) transport due to the uncoupling of Ca(2+)-stimulated ATP hydrolysis and ATP-driven Ca(2+) flux, and the downregulation of both sarco-endoplasmic reticulum Ca(2+)-ATPase 2 and the coupling factor 12-kDa FK506-binding protein. An increase in protein carbonylation (a marker of oxidative damage) and an imbalance between protein kinases C and A were observed as a legacy of undernutrition in early life. These results provide the structural and molecular basis to explain at least in part how maternal undernutrition affects fecundity and fertility in adult male rats.

15.
Am J Physiol Renal Physiol ; 309(10): F880-8, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26268270

RESUMEN

In contrast to the negative feedback of angiotensin II (ANG II) on juxtaglomerular renin, ANG II stimulates renin in the principal cells of the collecting duct (CD) in rats and mice via ANG II type 1 (AT1R) receptor, independently of blood pressure. In vitro data indicate that CD renin is augmented by AT1R activation through protein kinase C (PKC), but the exact mechanisms are unknown. We hypothesize that ANG II stimulates CD renin synthesis through AT1R via PKC and the subsequent activation of cAMP/PKA/CREB pathway. In M-1 cells, ANG II increased cAMP, renin mRNA (3.5-fold), prorenin, and renin proteins, as well as renin activity in culture media (2-fold). These effects were prevented by PKC inhibition with calphostin C, PKC-α dominant negative, and by PKA inhibition. Forskolin-induced increases in cAMP and renin expression were prevented by calphostin C. PKC inhibition and Ca2+ depletion impaired ANG II-mediated CREB phosphorylation and upregulation of renin. Adenylate cyclase 6 (AC) siRNA remarkably attenuated the ANG II-dependent upregulation of renin mRNA. Physiological activation of AC with vasopressin increased renin expression in M-1 cells. The results suggest that the ANG II-dependent upregulation of renin in the CD depends on PKC-α, which allows the augmentation of cAMP production and activation of PKA/CREB pathway via AC6. This study defines the intracellular signaling pathway involved in the ANG II-mediated stimulation of renin in the CD. This is a novel mechanism responsible for the regulation of local renin-angiotensin system in the distal nephron.


Asunto(s)
Angiotensina II/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína Quinasa C-alfa/metabolismo , Renina/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Ratones , Fosforilación , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos
16.
World J Nephrol ; 3(3): 64-76, 2014 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25332897

RESUMEN

The renin-angiotensin system (RAS) has been known for more than a century as a cascade that regulates body fluid balance and blood pressure. Angiotensin II(Ang II) has many functions in different tissues; however it is on the kidney that this peptide exerts its main functions. New enzymes, alternative routes for Ang IIformation or even active Ang II-derived peptides have now been described acting on Ang II AT1 or AT2 receptors, or in receptors which have recently been cloned, such as Mas and AT4. Another interesting observation was that old members of the RAS, such as angiotensin converting enzyme (ACE), renin and prorenin, well known by its enzymatic activity, can also activate intracellular signaling pathways, acting as an outside-in signal transduction molecule or on the renin/(Pro)renin receptor. Moreover, the endocrine RAS, now is also known to have paracrine, autocrine and intracrine action on different tissues, expressing necessary components for local Ang II formation. This in situ formation, especially in the kidney, increases Ang II levels to regulate blood pressure and renal functions. These discoveries, such as the ACE2/Ang-(1-7)/Mas axis and its antangonistic effect rather than classical deleterious Ang II effects, improves the development of new drugs for treating hypertension and cardiovascular diseases.

17.
Biochim Biophys Acta ; 1842(12 Pt A): 2357-66, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25283821

RESUMEN

This study has investigated the participation of altered signaling linked to angiotensin II (Ang II) that could be associated with increased Na(+) reabsorption in renal proximal tubules during chronic undernutrition. A multideficient chow for rats (basic regional diet, BRD) was used, which mimics several human diets widely taken in developing countries. The Vmax of the ouabain-resistant Na(+)-ATPase resident in the basolateral membranes increased >3-fold (P<0.001) accompanied by an increase in Na(+) affinity from 4.0 to 0.2mM (P<0.001). BRD rats had a >3-fold acceleration of the formation of phosphorylated intermediates in the early stage of the catalytic cycle (in the E1 conformation) (P<0.001). Immunostaining showed a huge increase in Ang II-positive cells in the cortical tubulointerstitium neighboring the basolateral membranes (>6-fold, P<0.001). PKC isoforms (α, ε, λ, ζ), Ang II type 1 receptors and PP2A were upregulated in BRD rats (in %): 55 (P<0.001); 35 (P<0.01); 125, 55, 11 and 30 (P<0.001). PKA was downregulated by 55% (P<0.001). With NetPhosK 1.0 and NetPhos 2.0, we detected 4 high-score (>0.70) regulatory phosphorylation sites for PKC and 1 for PKA in the primary sequence of the Na(+)-ATPase α-subunit, which are located in domains that are key for Na(+) binding and catalysis. Therefore, chronic undernutrition stimulates tubulointerstitial activity of Ang II and impairs PKC- and PKA-mediated regulatory phosphorylation, which culminates in an exaggerated Na(+) reabsorption across the proximal tubular epithelium.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Angiotensina II/metabolismo , Proteínas de Transporte de Catión/metabolismo , Riñón/enzimología , Desnutrición/fisiopatología , Transducción de Señal , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Angiotensina II/farmacología , Animales , Biocatálisis/efectos de los fármacos , Western Blotting , Proteínas de Transporte de Catión/química , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Furosemida/farmacología , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Cinética , Masculino , Desnutrición/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Ouabaína/farmacología , Fosforilación , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , Ratas Wistar , Receptor de Angiotensina Tipo 1/metabolismo , Sodio/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
PLoS One ; 9(7): e100410, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24983243

RESUMEN

BACKGROUND: Several studies have correlated protein restriction associated with other nutritional deficiencies with the development of cardiovascular and renal diseases. The driving hypothesis for this study was that Ang II signaling pathways in the heart and kidney are affected by chronic protein, mineral and vitamin restriction. METHODOLOGY/PRINCIPAL FINDINGS: Wistar rats aged 90 days were fed from weaning with either a control or a deficient diet that mimics those used in impoverished regions worldwide. Such restriction simultaneously increased ouabain-insensitive Na+-ATPase and decreased (Na++K+)ATPase activity in the same proportion in cardiomyocytes and proximal tubule cells. Type 1 angiotensin II receptor (AT1R) was downregulated by that restriction in both organs, whereas AT2R decreased only in the kidney. The PKC/PKA ratio increased in both tissues and returned to normal values in rats receiving Losartan daily from weaning. Inhibition of the MAPK pathway restored Na+-ATPase activity in both organs. The undernourished rats presented expanded plasma volume, increased heart rate, cardiac hypertrophy, and elevated systolic pressure, which also returned to control levels with Losartan. Such restriction led to electrical cardiac remodeling represented by prolonged ventricular repolarization parameters, induced triggered activity, early after-depolarization and delayed after-depolarization, which were also prevented by Losartan. CONCLUSION/SIGNIFICANCE: The mechanisms responsible for these alterations are underpinned by an imbalance in the PKC- and PKA-mediated pathways, with participation of angiotensin receptors and by activation of the MAPK/ERK1/2 pathway. These cellular and molecular alterations culminate in cardiac electric remodeling and in the onset of hypertension in adulthood.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Desnutrición/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Adenosina Trifosfatasas/metabolismo , Angiotensina II , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Presión Sanguínea , Cardiomegalia/patología , Proteínas de Transporte de Catión/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Corazón/fisiopatología , Frecuencia Cardíaca , Túbulos Renales Proximales/metabolismo , Losartán/farmacología , Masculino , Miocitos Cardíacos/metabolismo , Volumen Plasmático , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
19.
Am J Physiol Renal Physiol ; 306(8): F855-63, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24523384

RESUMEN

The physiological roles of ANG-(3-4) (Val-Tyr), a potent ANG II-derived peptide, remain largely unknown. The present study 1)investigates whether ANG-(3-4) modulates ouabain-resistant Na(+)-ATPase resident in proximal tubule cells and 2) verifies whether its possible action on pumping activity, considered the fine tuner of Na(+) reabsorption in this nephron segment, depends on blood pressure. ANG-(3-4) inhibited Na(+)-ATPase activity in membranes of spontaneously hypertensive rats (SHR) at nanomolar concentrations, with no effect in Wistar-Kyoto (WKY) rats or on Na(+)-K(+)-ATPase. PD123319 (10(-7) M) and PKA(5-24) (10(-6) M), an AT2 receptor (AT2R) antagonist and a specific PKA inhibitor, respectively, abrogated this inhibition, indicating that AT2R and PKA are central in this pathway. Despite the lack of effect of ANG-(3-4) when assayed alone in WKY rats, the peptide (10(-8) M) completely blocked stimulation of Na(+)-ATPase induced by 10(-10) M ANG II in normotensive rats through a mechanism that also involves AT2R and PKA. Tubular membranes from WKY rats had higher levels of AT2R/AT1R heterodimers, which remain associated in 10(-10) M ANG II and dissociate to a very low dimerization state upon addition of 10(-8) M ANG-(3-4). This lower level of heterodimers was that found in SHR, and heterodimers did not dissociate when the same concentration of ANG-(3-4) was present. Oral administration of ANG-(3-4) (50 mg/kg body mass) increased urinary Na(+) concentration and urinary Na(+) excretion with a simultaneous decrease in systolic arterial pressure in SHR, but not in WKY rats. Thus the influence of ANG-(3-4) on Na(+) transport and its hypotensive action depend on receptor association and on blood pressure.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Proteínas de Transporte de Catión/antagonistas & inhibidores , Dipéptidos/farmacología , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Hipertensión/fisiopatología , Imidazoles/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Ouabaína/farmacología , Piridinas/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 2/efectos de los fármacos , Receptor de Angiotensina Tipo 2/fisiología , Sodio/orina , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
20.
PLoS One ; 8(7): e69682, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23922775

RESUMEN

BACKGROUND: The aim of this work was to investigate the mechanisms by which chronic malnutrition (CM) affects vas deferens function, leading to compromised reproductive capacity. Previous studies have shown that maternal malnutrition affects the reproductive tracts of adult male offspring. However, little is known about the effects of CM, a widespread life-long condition that persists from conception throughout growth to adult life. METHODOLOGY/PRINCIPAL FINDINGS: Young adult male rats, which were chronically malnourished from weaning, presented decreased total and haploid cells in the vas deferens, hypertrophy of the muscle layer in the epididymal portion of the vas deferens and intense atrophy of the muscular coat in its prostatic portion. At a molecular level, the vas deferens tissue of CM rats exhibited a huge rise in lipid peroxidation and protein carbonylation, evidence of an accentuated increase in local reactive oxygen species levels. The kinetics of plasma membrane Ca(2+)-ATPase activity and its kinase-mediated phosphorylation by PKA and PKC in the vas deferens revealed malnutrition-induced modifications in velocity, Ca(2+) affinity and regulation of Ca(2+) handling proteins. The severely crippled content of the 12-kDa FK506 binding protein, which controls passive Ca(2+) release from the sarco(endo) plasmic reticulum, revealed another target of malnutrition related to intracellular Ca(2+) handling, with a potential effect on forward propulsion of sperm cells. As a possible compensatory response, malnutrition led to enhanced sarco(endo) plasmic reticulum Ca(2+)-ATPase activity, possibly caused by stimulatory PKA-mediated phosphorylation. CONCLUSIONS/SIGNIFICANCE: The functional correlates of these cellular and molecular hallmarks of chronic malnutrition on the vas deferens were an accentuated reduction in fertility and fecundity.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Desnutrición/patología , Estrés Oxidativo , Reproducción , Conducto Deferente/metabolismo , Conducto Deferente/patología , Envejecimiento/patología , Animales , Transporte Biológico , Peso Corporal , ATPasas Transportadoras de Calcio/metabolismo , Recuento de Células , Supervivencia Celular , Enfermedad Crónica , Epidídimo/patología , Haploidia , Cinética , Masculino , Desnutrición/enzimología , Músculos/patología , Tamaño de los Órganos , Oxidación-Reducción , Fosforilación , Ratas , Ratas Wistar , Espermatozoides/patología , Testículo/patología , Conducto Deferente/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA