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

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(9)2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37175599

RESUMO

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.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão , Ratos , Masculino , Animais , Cloreto de Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Ratos Wistar , Hipertensão/metabolismo , Sódio/metabolismo , Cloreto de Sódio na Dieta/efeitos adversos , Cloreto de Sódio na Dieta/metabolismo , Pressão Sanguínea , Rim/metabolismo , Íons/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
2.
Nutr Neurosci ; 25(9): 1973-1994, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33871318

RESUMO

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.


Assuntos
Desnutrição , Animais , Brasil , Dieta , Comportamento Alimentar , Desnutrição/epidemiologia
3.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 102-114, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28987762

RESUMO

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.


Assuntos
Injúria Renal Aguda/patologia , Citoproteção/efeitos dos fármacos , Doxiciclina/farmacologia , Rim/efeitos dos fármacos , Traumatismo por Reperfusão/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Relação Dose-Resposta a Droga , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/irrigação sanguínea , Rim/patologia , Rim/fisiologia , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/fisiopatologia
4.
Am J Physiol Renal Physiol ; 313(2): F440-F449, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28468964

RESUMO

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.


Assuntos
Angiotensina II/metabolismo , Membrana Celular/metabolismo , Endocitose , Retículo Endoplasmático/metabolismo , Rim/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Rim/efeitos dos fármacos , Células LLC-PK1 , Microtúbulos/metabolismo , Complexos Multiproteicos , Fosforilação , Proteína Quinase C-alfa/metabolismo , Transporte Proteico , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 2 de Angiotensina/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Suínos , beta-Arrestinas/metabolismo
5.
Curr Hypertens Rep ; 19(8): 62, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28695400

RESUMO

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.


Assuntos
Hipertensão/metabolismo , Hipertensão/fisiopatologia , Túbulos Renais Coletores/metabolismo , Renina/metabolismo , Angiotensina II/metabolismo , Animais , Humanos , Hipertensão/etiologia , Túbulos Renais Coletores/fisiopatologia , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/fisiopatologia , Túbulos Renais Proximais/fisiopatologia , Renina/biossíntese , Sistema Renina-Angiotensina/fisiologia
6.
Lipids Health Dis ; 16(1): 245, 2017 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-29246161

RESUMO

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.


Assuntos
Colesterol/metabolismo , Hipertensão/metabolismo , Rim/metabolismo , Desnutrição/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Insuficiência Renal Crônica/metabolismo , 1-Fosfatidilinositol 4-Quinase/genética , 1-Fosfatidilinositol 4-Quinase/metabolismo , Animais , Animais Recém-Nascidos , Ceramidas/metabolismo , Diacilglicerol Quinase/genética , Diacilglicerol Quinase/metabolismo , Regulação da Expressão Gênica , Hipertensão/etiologia , Hipertensão/genética , Hipertensão/patologia , Rim/química , Metabolismo dos Lipídeos , Masculino , Desnutrição/complicações , Desnutrição/genética , Desnutrição/patologia , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Ácidos Fosfatídicos/metabolismo , Fosfolipases A2/genética , Fosfolipases A2/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ratos , Ratos Wistar , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia
7.
Biochim Biophys Acta ; 1842(12 Pt A): 2357-66, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25283821

RESUMO

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.


Assuntos
Adenosina Trifosfatases/metabolismo , Angiotensina II/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Rim/enzimologia , Desnutrição/fisiopatologia , Transdução de Sinais , Adenosina Trifosfatases/química , Sequência de Aminoácidos , Angiotensina II/farmacologia , Animais , Biocatálise/efeitos dos fármacos , Western Blotting , Proteínas de Transporte de Cátions/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Furosemida/farmacologia , Humanos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Cinética , Masculino , Desnutrição/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ouabaína/farmacologia , Fosforilação , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Ratos Wistar , Receptor Tipo 1 de Angiotensina/metabolismo , Sódio/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Am J Physiol Renal Physiol ; 309(10): F880-8, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26268270

RESUMO

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.


Assuntos
Angiotensina II/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína Quinase C-alfa/metabolismo , Renina/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Camundongos , Fosforilação , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos
9.
Am J Physiol Renal Physiol ; 306(8): F855-63, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24523384

RESUMO

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.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Proteínas de Transporte de Cátions/antagonistas & inibidores , Dipeptídeos/farmacologia , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Hipertensão/fisiopatologia , Imidazóis/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Ouabaína/farmacologia , Piridinas/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptor Tipo 2 de Angiotensina/efeitos dos fármacos , Receptor Tipo 2 de Angiotensina/fisiologia , Sódio/urina , ATPase Trocadora de Sódio-Potássio/metabolismo
10.
Am J Physiol Renal Physiol ; 302(1): F85-94, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21900456

RESUMO

Augmentation of intrarenal angiotensinogen (AGT) synthesis, secretion, and excretion is associated with the development of hypertension, renal oxidative stress, and tissue injury during ANG II-dependent hypertension. High salt (HS) exacerbates hypertension and kidney injury, but the mechanisms remain unclear. In this study, we determined the consequences of HS intake alone compared with chronic ANG II infusion and combined HS plus ANG II on the stimulation of urinary AGT (uAGT), renal oxidative stress, and renal injury markers. Sprague-Dawley rats were subjected to 1) a normal-salt diet [NS, n = 5]; 2) HS diet [8% NaCl, n = 5]; 3) ANG II infusion in NS rats [ANG II 80 ng/min, n = 5]; 4) ANG II infusion in HS rats [ANG II+HS, n = 5]; and 5) ANG II infusion in HS rats treated with ANG II type 1 receptor blocker (ARB) [ANG II+HS+ARB, n = 5] for 14 days. Rats fed a HS diet alone did not show changes in systolic blood pressure (SBP), proteinuria, cell proliferation, or uAGT excretion although they did exhibit mesangial expansion, collagen deposition, and had increased NADPH oxidase activity accompanied by increased peroxynitrite formation in the kidneys. Compared with ANG II rats, the combination of ANG II infusion and a HS diet led to exacerbation in SBP (175 ± 10 vs. 221 ± 8 mmHg; P < 0.05), proteinuria (46 ± 7 vs. 127 ± 7 mg/day; P < 0.05), and uAGT (1,109 ± 70 vs.. 7,200 ± 614 ng/day; P < 0.05) associated with greater collagen deposition, mesangial expansion, interstitial cell proliferation, and macrophage infiltration. In both ANG II groups, the O(2)(-) levels were increased due to increased NADPH oxidase activity without concomitant increases in peroxynitrite formation. The responses in ANG II rats were prevented or ameliorated by ARB treatment. The results indicate that HS independently stimulates ROS formation, which may synergize with the effect of ANG II to limit peroxynitrite formation, leading to exacerbation of uAGT and greater injury during ANG II salt hypertension.


Assuntos
Angiotensinogênio/biossíntese , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/fisiologia , Cloreto de Sódio na Dieta/administração & dosagem , Angiotensina II , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Angiotensinogênio/urina , Animais , Hipertensão/induzido quimicamente , Hipertensão/patologia , Rim/patologia , Masculino , NADPH Oxidases/metabolismo , Ácido Peroxinitroso/biossíntese , Proteinúria/etiologia , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio na Dieta/farmacologia
11.
Am J Physiol Renal Physiol ; 303(1): F105-9, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22442212

RESUMO

Increased dietary salt triggers oxidative stress and kidney injury in salt-sensitive hypertension; however, the mechanism for sensing increased extracellular Na(+) concentration ([Na(+)]) remains unclear. A Na(+)-activated Na(+) channel (Na sensor) described in the brain operates as a sensor of extracellular fluid [Na(+)]; nonetheless, its presence in the kidney has not been established. In the present study, we demonstrated the gene expression of the Na sensor by RT-PCR and Western blotting in the Sprague-Dawley rat kidney. Using immunofluorescence, the Na sensor was localized to the luminal side in tubular epithelial cells of collecting ducts colocalizing with aquaporin-2, a marker of principal cells, and in thick ascending limb, colocalizing with the glycoprotein Tamm-Horsfall. To determine the effect of a high-salt diet (HSD) on Na sensor gene expression, we quantified its transcript and protein levels primarily in renal medullas from control rats and rats subjected to 8% NaCl for 7 days (n = 5). HSD increased Na sensor expression levels (mRNA: from 1.2 ± 0.2 to 5.1 ± 1.3 au; protein: from 0.98 ± 0.15 to 1.74 ± 0.28 au P < 0.05) in the kidney medulla, but not in the cortex. These data indicate that rat kidney epithelial cells of the thick ascending limb and principal cells of the collecting duct possess a Na sensor that is upregulated by HSD, suggesting an important role in monitoring changes in tubular fluid [Na(+)].


Assuntos
Células Epiteliais/metabolismo , Rim/metabolismo , Canais de Sódio/metabolismo , Cloreto de Sódio na Dieta/administração & dosagem , Animais , Aquaporina 2/genética , Aquaporina 2/metabolismo , Células Epiteliais/efeitos dos fármacos , Expressão Gênica , Rim/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Canais de Sódio/genética , Regulação para Cima
12.
Am J Physiol Renal Physiol ; 302(7): F875-83, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22218590

RESUMO

ANG II is secreted into the lumens of proximal tubules where it is also synthesized, thus increasing the local concentration of the peptide to levels of potential physiological relevance. In the present work, we studied the effect of ANG II via the luminal membranes of LLC-PK(1) cells on Ca(2+)-ATPase of the sarco(endo)plasmic reticulum (SERCA) and plasma membrane (PMCA). ANG II (at concentrations found in the lumen) stimulated rapid (30 s) and persistent (30 min) SERCA activity by more than 100% and increased Ca(2+) mobilization. Pretreatment with ANG II for 30 min enhanced the ANG II-induced Ca(2+) spark, demonstrating a positively self-sustained stimulus of Ca(2+) mobilization by ANG II. ANG II in the medium facing the luminal side of the cells decreased with time with no formation of metabolites, indicating peptide internalization. ANG II increased heterodimerization of AT(1) and AT(2) receptors by 140%, and either losartan or PD123319 completely blocked the stimulation of SERCA by ANG II. Using the PLC inhibitor U73122, PMA, and calphostin C, it was possible to demonstrate the involvement of a PLC→DAG(PMA)→PKC pathway in the stimulation of SERCA by ANG II with no effect on PMCA. We conclude that ANG II triggers SERCA activation via the luminal membrane, increasing the Ca(2+) stock in the reticulum to ensure a more efficient subsequent mobilization of Ca(2+). This first report on the regulation of SERCA activity by ANG II shows a new mechanism for Ca(2+) homeostasis in renal cells and also for regulation of Ca(2+)-modulated fluid reabsorption in proximal tubules.


Assuntos
Angiotensina II/metabolismo , Túbulos Renais Proximais/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/enzimologia , Neprilisina/metabolismo , Peptídeo Hidrolases/metabolismo , Peptidil Dipeptidase A/metabolismo , Multimerização Proteica , Transdução de Sinais , Suínos
13.
Curr Hypertens Rev ; 18(2): 91-100, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35170417

RESUMO

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.


Assuntos
Hipertensão , Renina , Humanos , Angiotensina II , Pressão Sanguínea , Receptor de Pró-Renina , Receptores de Superfície Celular/metabolismo , Sistema Renina-Angiotensina
14.
Biochim Biophys Acta ; 1798(3): 360-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19560439

RESUMO

Clinical and experimental data show an increase in sodium reabsorption on the proximal tubule (PT) in essential hypertension. It is well known that there is a link between essential hypertension and renal angiotensin II (Ang II). The present study was designed to examine ouabain-insensitive Na(+)-ATPase activity and its regulation by Ang II in spontaneously hypertensive rats (SHR). We observed that Na(+)-ATPase activity was enhanced in 14-week-old but not in 6-week-old SHR. The addition of Ang II from 10(-12) to 10(-6) mol/L decreased the enzyme activity in SHR to a level similar to that obtained in WKY. The Ang II inhibitory effect was completely reversed by a specific antagonist of AT(2) receptor, PD123319 (10(-8) mol/L) indicating that a system leading to activation of the enzyme in SHR is inhibited by AT(2)-mediated Ang II. Treatment of SHR with losartan for 10 weeks (weeks 4-14) prevents the increase in Na(+)-ATPase activity observed in 14-week-old SHR. These results indicate a correlation between AT(1) receptor activation in SHR and increased ouabain-insensitive Na(+)-ATPase activity. Our results open new possibilities towards our understanding of the pathophysiological mechanisms involved in the increased sodium reabsorption in PT found in essential hypertension.


Assuntos
Hipertensão/enzimologia , Hipertensão/patologia , Receptor Tipo 1 de Angiotensina/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Angiotensina II/farmacologia , Animais , Ativação Enzimática/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Cinética , Losartan/farmacologia , Masculino , Ouabaína/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
15.
Am J Physiol Renal Physiol ; 300(3): F749-55, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21209009

RESUMO

Alterations in the balance between ANG II/ACE and ANG 1-7/ACE2 in ANG II-dependent hypertension could reduce the generation of ANG 1-7 and contribute further to increased intrarenal ANG II. Upregulation of collecting duct (CD) renin may lead to increased ANG II formation during ANG II-dependent hypertension, thus contributing to this imbalance. We measured ANG I, ANG II, and ANG 1-7 contents, angiotensin-converting enzyme (ACE) and ACE2 gene expression, and renin activity in the renal cortex and medulla in the clipped kidneys (CK) and nonclipped kidneys (NCK) of 2K1C rats. After 3 wk of unilateral renal clipping, systolic blood pressure and plasma renin activity increased in 2K1C rats (n = 11) compared with sham rats (n = 9). Renal medullary angiotensin peptide levels were increased in 2K1C rats [ANG I: (CK = 171 ± 4; NCK = 251 ± 8 vs. sham = 55 ± 3 pg/g protein; P < 0.05); ANG II: (CK = 558 ± 79; NCK = 328 ± 18 vs. sham = 94 ± 7 pg/g protein; P < 0.001)]; and ANG 1-7 levels decreased (CK = 18 ± 2; NCK = 19 ± 2 pg/g vs. sham = 63 ± 10 pg/g; P < 0.001). In renal medullas of both kidneys of 2K1C rats, ACE mRNA levels and activity increased but ACE2 decreased. In further studies, we compared renal ACE and ACE2 mRNA levels and their activities from chronic ANG II-infused (n = 6) and sham-operated rats (n = 5). Although the ACE mRNA levels did not differ between ANG II rats and sham rats, the ANG II rats exhibited greater ACE activity and reduced ACE2 mRNA levels and activity. Renal medullary renin activity was similar in the CK and NCK of 2K1C rats but higher compared with sham. Thus, the differential regulation of ACE and ACE2 along with the upregulation of CD renin in both the CK and NCK in 2K1C hypertensive rats indicates that they are independent of perfusion pressure and contribute to the altered content of intrarenal ANG II and ANG 1-7.


Assuntos
Angiotensina II/metabolismo , Angiotensina I/metabolismo , Hipertensão Renovascular/metabolismo , Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/metabolismo , Renina/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Córtex Renal/metabolismo , Medula Renal/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos
16.
Arch Biochem Biophys ; 505(1): 91-7, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20887709

RESUMO

Prenatal malnutrition is responsible for the onset of alterations in renal Na(+) transport in the adult offspring. Here we investigated the molecular mechanisms by which increased formation of reactive oxygen species during prenatal malnutrition affects the pathways that couple angiotensin II (Ang II) receptors (AT(1)R and AT(2)R) to kidney Na(+)-ATPase in adulthood, and how maternal treatment with α-tocopherol can prevent alterations in the main regulatory cascade of the pump. The experiments were carried out on the adult progeny of control and malnourished dams during pregnancy that did or did not receive α-tocopherol during lactation. Malnutrition during pregnancy increased maternal hepatic and adult offspring renal malondialdehyde levels, which returned to control after supplementation with α-tocopherol. In the adult offspring, placental malnutrition programmed: decrease in Na(+)-ATPase activity, loss of the physiological stimulation of this pump by Ang II, up-regulation of AT(1)R and AT(2)R, decrease in membrane PKC activity, selective decrease of the PKCε isoform expression, and increase in PKA activity with no change in PKA α-catalytic subunit expression. These alterations were reprogrammed to normal levels by α-tocopherol during lactation. The influence of α-tocopherol on the signaling machinery in adult offspring indicates selective non-antioxidant effects at the gene transcription and protein synthesis levels.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Rim/enzimologia , Desnutrição/complicações , Placenta/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Angiotensina/metabolismo , alfa-Tocoferol/uso terapêutico , Animais , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/metabolismo , Lactação , Desnutrição/metabolismo , Gravidez , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptores de Angiotensina/genética , alfa-Tocoferol/administração & dosagem
17.
Toxicon ; 199: 117-126, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34116084

RESUMO

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.


Assuntos
Injúria Renal Aguda , Bothrops , Venenos de Crotalídeos , Injúria Renal Aguda/induzido quimicamente , Animais , Venenos de Crotalídeos/toxicidade , Taxa de Filtração Glomerular , Rim , Masculino , Ratos , Ratos Wistar
18.
Toxicon ; 199: 20-30, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34058237

RESUMO

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.


Assuntos
Bothrops , Venenos de Crotalídeos , Animais , Venenos de Crotalídeos/toxicidade , Doxiciclina , Humanos , Rim/fisiologia , Masculino , Ratos , Ratos Wistar
19.
Arch Biochem Biophys ; 496(2): 117-22, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20153712

RESUMO

We showed previously that angiotensin-(1-7) [Ang-(1-7)] reversed stimulation of proximal tubule Na+-ATPase promoted by angiotensin II (Ang II) through a D-ala(7)-Ang-(1-7) (A779)-sensitive receptor. Here we investigated the signaling pathway coupled to this receptor. According to our data, Ang-(1-7) produces a MAS-mediated reversal of Ang II-stimulated Na+-ATPase by a Gs/PKA pathway because: (1) the Ang-(1-7) effect is reversed by GDPbetaS, an inhibitor of trimeric G protein and Gs polyclonal antibody. Cholera toxin, an activator of Gs protein, mimicked it; (2) in the presence of Ang II, Ang-(1-7) increased the PKA activity 10-fold; (3) the peptide inhibitor of PKA blocked the Ang-(1-7) effect on Ang II-stimulated Na+-ATPase; (4) Ang-(1-7) reverses the Ang II-stimulated PKC activity; (5) cAMP mimicked the Ang-(1-7) effect on the Ang II-stimulated Na+-ATPase. Our results provide new understanding about the signaling mechanisms coupled to MAS receptor-mediated renal Ang-(1-7) effects.


Assuntos
Angiotensina II/administração & dosagem , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Sódio/metabolismo , Animais , Células Cultivadas , Proto-Oncogene Mas , Suínos
20.
Nephrol Dial Transplant ; 25(12): 3867-74, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20504839

RESUMO

BACKGROUND: Bioactive lipids are important in tissue injury and regeneration. Ceramide (Cer) is known for its pro-apoptotic action and sphingosine-1-phosphate (S1P) for inducing proliferation and cell survival; diacylglycerol (DAG) and lysophosphatidic acid (LPA) are involved in various signalling pathways including modulation of ion transport. LPA signalling through its receptor LPA(1) is also related to the progression of fibrosis. This study investigated the modulation of lipid signalling pathways induced by administration of bone marrow-derived mononuclear cells (BMMC) in chronic kidney disease. METHODS: Unilateral ureteral obstruction (UUO) was followed by intravenous injection of ∼2 × 10(7) BMMC. Controls were UUO group treated with buffered solution and sham-operated group. Animals were killed 14 days after surgery, and lipid phosphorylation assays and immunoblotting were performed on the kidney homogenates. RESULTS: More DAG was available in the UUO rats (2.4 ± 0.4 and 2.4 ± 0.3 vs 1.0 ± 0.2 pmol (32)PA mg(-)(1) min(-)(1), in UUO and UUO + BMMC vs SHAM). Sphingosine kinase was 150 ± 12% more active in UUO + BMMC than in UUO and SHAM. Cer levels were 76 ± 7% lower in the UUO + BMMC than UUO. LPA receptor type 1 (LPA(1)) expression was 169 ± 7% higher in the UUO group than in UUO + BMMC and SHAM. BMMC maintain control levels of Ca(2+)-ATPase expression altered by UUO by 40%. CONCLUSIONS: BMMC infusion modulated diverse lipid signalling pathways and protein expression, shifted sphingolipid metabolism toward a regenerative pattern and favourably reduced the levels of a receptor involved in the progression of tissue fibrosis. These results strengthen the benefits of BMMC treatment and give insight into its paracrine mechanisms of action.


Assuntos
Injúria Renal Aguda/fisiopatologia , Células da Medula Óssea/fisiologia , Rim/fisiologia , Lipídeos/fisiologia , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Obstrução Ureteral/fisiopatologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , ATPases Transportadoras de Cálcio/metabolismo , Proliferação de Células , Sobrevivência Celular , Diglicerídeos/metabolismo , Rim/patologia , Masculino , Modelos Animais , Ratos , Ratos Wistar , Receptores de Ácidos Lisofosfatídicos/metabolismo , Esfingolipídeos/metabolismo , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA