Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 323(1): F33-F47, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35532070

RESUMO

Ischemia-reperfusion (I/R) is considered the primary cause of acute kidney injury and is higher among older individuals. Although ischemic episodes are hard to predict and prevent, detrimental ischemic effects could be mitigated by exogenous intervention. This study aimed to identify the protective role of angiotensin (1-7) [ANG(1-7)] against I/R-induced renal injury in adult versus aged rats. Adult and aged male Fisher 344 rats were subjected to 40 min of bilateral renal ischemia followed by 28 days of reperfusion. ANG(1-7) was administered intraperitoneally in ischemic rats for 28 days with or without the Mas receptor antagonist A779. I/R increased blood pressure, plasma creatinine, urinary 8-isoprostane, and renal infiltration of pro- and anti-inflammatory macrophages and reduced glomerular filtration rate in both adult and aged rats compared with sham rats. In addition to causing glomerular sclerosis and tubular damage, I/R increased the expression of the following pathogenic miRNAs: miR-20a-5p, miR-21-5p, miR-24-3p, and miR-194-5p in both age groups. ANG(1-7) treatment of ischemic rats mitigated oxidative stress and renal inflammation, restored renal structure and function, and reduced high blood pressure. Also, ANG(1-7) suppressed the expression of pathogenic miRNAs. In addition, ANG(1-7) treatment of I/R rats increased the expression of the redox-sensitive transcription factor nuclear factor-erythroid factor 2-related factor 2 (NRF2) and phase II antioxidant enzymes. The beneficial effects of ANG(1-7) were sensitive to A779. Collectively, these data suggest that ANG(1-7) associated with NRF2 activation could alleviate post-I/R-induced kidney injury, and therefore serve as a potential therapeutic compound to protect against biochemical and morphological pathologies of I/R in both adults and aged populations.NEW & NOTEWORTHY This is the first study to show that ANG(1-7) via Mas receptors could activate the redox-sensitive nuclear factor-erythroid factor 2-related factor 2 (NRF2)-phase II antioxidant system and protect against ischemia-reperfusion (I/R)-induced renal injury, identifying ANG(1-7), Mas receptor agonists, or NRF2 activators as potential therapeutic interventions to protect against renal and cardiovascular diseases. Moreover, miRNAs have differential expression in adult versus aged rats, and I/R modulates miRNA expression in both age groups, indicating their involvement in kidney disease.


Assuntos
Injúria Renal Aguda , MicroRNAs , Traumatismo por Reperfusão , Injúria Renal Aguda/patologia , Angiotensina I , Animais , Antioxidantes/farmacologia , Isquemia/metabolismo , Rim/metabolismo , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fragmentos de Peptídeos , Ratos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle
2.
Antioxidants (Basel) ; 11(1)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35052660

RESUMO

Age is one of the major risk factors for the development of chronic pathologies, including kidney diseases. Oxidative stress and mitochondrial dysfunction play a pathogenic role in aging kidney disease. Transcription factor NRF2, a master regulator of redox homeostasis, is altered during aging, but the exact implications of altered NRF2 signaling on age-related renal mitochondrial impairment are not yet clear. Herein, we investigated the role of sulforaphane, a well-known NRF2 activator, on age-related mitochondrial and kidney dysfunction. Young (2-4 month) and aged (20-24 month) male Fischer 344 rats were treated with sulforaphane (15 mg/kg body wt/day) in drinking water for four weeks. We observed significant impairment in renal cortical mitochondrial function along with perturbed redox homeostasis, decreased kidney function and marked impairment in NRF2 signaling in aged Fischer 344 rats. Sulforaphane significantly improved mitochondrial function and ameliorated kidney injury by increasing cortical NRF2 expression and activity and decreasing protein expression of KEAP1, an NRF2 repressor. Sulforaphane treatment did not affect the renal NRF2 expression or activity and mitochondrial function in young rats. Taken together, our results provide novel insights into the protective role of the NRF2 pathway in kidneys during aging and highlight the therapeutic potential of sulforaphane in mitigating kidney dysfunction in elders.

3.
J Am Heart Assoc ; 9(1): e014977, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31880979

RESUMO

Background Oxidative stress and high salt intake could be independent or intertwined risk factors in the origin of hypertension. Kidneys are the major organ to regulate sodium homeostasis and blood pressure and the renal dopamine system plays a pivotal role in sodium regulation during sodium replete conditions. Oxidative stress has been implicated in renal dopamine dysfunction and development of hypertension, especially in salt-sensitive animal models. Here we show the nexus between high salt intake and oxidative stress causing renal tubular dopamine oxidation, which leads to mitochondrial and lysosomal dysfunction and subsequently causes renal inflammation and hypertension. Methods and Results Male Sprague Dawley rats were divided into the following groups, vehicle (V)-tap water, high salt (HS)-1% NaCl, L-buthionine-sulfoximine (BSO), a prooxidant, and HS plus BSO without and with antioxidant resveratrol (R) for 6 weeks. Oxidative stress was significantly higher in BSO and HS+BSO-treated rat compared with vehicle; however, blood pressure was markedly higher in the HS+BSO group whereas an increase in blood pressure in the BSO group was modest. HS+BSO-treated rats had significant renal dopamine oxidation, lysosomal and mitochondrial dysfunction, and increased renal inflammation; however, HS alone had no impact on organelle function or inflammation. Resveratrol prevented oxidative stress, dopamine oxidation, organelle dysfunction, inflammation, and hypertension in BSO and HS+BSO rats. Conclusions These data suggest that dopamine oxidation, especially during increased sodium intake and oxidative milieu, leads to lysosomal and mitochondrial dysfunction and renal inflammation with subsequent increase in blood pressure. Resveratrol, while preventing oxidative stress, protects renal function and mitigates hypertension.


Assuntos
Pressão Sanguínea , Dopamina/metabolismo , Hipertensão/etiologia , Túbulos Renais Proximais/metabolismo , Nefrite/etiologia , Estresse Oxidativo , Cloreto de Sódio na Dieta/toxicidade , Animais , Antioxidantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Lisossomos/metabolismo , Masculino , Mitocôndrias/metabolismo , Nefrite/metabolismo , Nefrite/fisiopatologia , Nefrite/prevenção & controle , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley
4.
Am J Physiol Renal Physiol ; 317(4): F949-F956, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31411069

RESUMO

The role of dopamine D1-like receptors (DR) in the regulation of renal Na+ transporters, natriuresis, and blood pressure is well established. However, the involvement of the angiotensin 1-7 (ANG 1-7)-Mas receptor in the regulation of Na+ balance and blood pressure is not clear. The present study aimed to investigate the hypothesis that ANG 1-7 can regulate Na+ homeostasis by modulating the renal dopamine system. Sprague-Dawley rats were infused with saline alone (vehicle) or saline with ANG 1-7, ANG 1-7 antagonist A-779, DR agonist SKF38393, and antagonist SCH23390. Infusion of ANG 1-7 caused significant natriuresis and diuresis compared with saline alone. Both natriuresis and diuresis were blocked by A-779 and SCH23390. SKF38393 caused a significant, SCH23390-sensitive natriuresis and diuresis, and A-779 had no effect on the SKF38393 response. Concomitant infusion of ANG 1-7 and SKF38393 did not show a cumulative effect compared with either agonist alone. Treatment of renal proximal tubules with ANG 1-7 or SKF38393 caused a significant decrease in Na+-K+-ATPase and Na+/H+ exchanger isoform 3 activity. While SCH23390 blocked both ANG 1-7- and SKF38393-induced inhibition, the DR response was not sensitive to A-779. Additionally, ANG 1-7 activated PKG, enhanced tyrosine hydroxylase activity via Ser40 phosphorylation, and increased renal dopamine production. These data suggest that ANG 1-7, via PKG, enhances tyrosine hydroxylase activity, which increases renal dopamine production and activation of DR and subsequent natriuresis. This study provides evidence for a unidirectional functional interaction between two G protein-coupled receptors to regulate renal Na+ transporters and induce natriuresis.


Assuntos
Angiotensina I/farmacologia , Rim/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptores de Dopamina D1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sódio/metabolismo , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Angiotensina I/antagonistas & inibidores , Animais , Benzazepinas/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Diurese/efeitos dos fármacos , Dopamina/biossíntese , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Natriurese/efeitos dos fármacos , Fragmentos de Peptídeos/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
5.
Clin Exp Hypertens ; 41(1): 5-13, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29424564

RESUMO

Reactive oxygen species induce vascular dysfunction and hypertension by directly interacting with nitric oxide (NO) which leads to NO inactivation. In addition to a decrease in NO bioavailability, there is evidence that oxidative stress can also modulate NO signaling during hypertension. Here, we investigated the effect of oxidative stress on NO signaling molecules cGMP-dependent protein kinase (PKG) and vasodilator-stimulated phosphoprotein (VASP) which are known to mediate vasodilatory actions of NO. Male Sprague Dawley (SD) rats were provided with tap water (control), 30 mM L-buthionine sulfoximine (BSO, a pro-oxidant), 1 mM tempol (T, an antioxidant) and BSO + T for 3 wks. BSO-treated rats exhibited high blood pressure and oxidative stress. Incubation of mesenteric arterial rings with NO donors caused concentration-dependent relaxation in control rats. However, the response to NO donors was significantly lower in BSO-treated rats with a marked decrease in pD2. In control rats, NO donors activated mesenteric PKG, increased VASP phosphorylation and its interaction with transient receptor potential channels 4 (TRPC4) and inhibited store-operated Ca2+ influx. NO failed to activate these signaling molecules in mesenteric arteries from BSO-treated rats. Supplementation of BSO-treated rats with tempol reduced oxidative stress and blood pressure and normalized the NO signaling. These data suggest that oxidative stress can reduce NO-mediated PKG activation and VASP-TRPC4 interaction which leads to failure of NO to reduce Ca2+ influx in smooth muscle cells. The increase in intracellular Ca2+ contributes to sustained vasoconstriction and subsequent hypertension. Antioxidant supplementation decreases oxidative stress, normalizes NO signaling and reduces blood pressure.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Hipertensão/fisiopatologia , Artérias Mesentéricas/fisiopatologia , Proteínas dos Microfilamentos/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Fosfoproteínas/metabolismo , Animais , Antioxidantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Cálcio/metabolismo , Óxidos N-Cíclicos/farmacologia , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/farmacologia , Serina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Marcadores de Spin , Canais de Cátion TRPC/metabolismo , Vasodilatação/efeitos dos fármacos
6.
Physiol Rep ; 3(11)2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26603454

RESUMO

Previously we have shown that in spontaneously hypertensive rats (SHR) renal angiotensin (Ang) II receptor (AT1R) upregulation leads to overstimulation of Na/K-ATPase by Ang II. There are reports that antioxidants can reduce oxidative stress and blood pressure (BP) in SHR, however the effect of these compounds on AT1R function remains to be determined. Therefore, we hypothesized that polyphenol antioxidant resveratrol would mitigate oxidative stress, normalize renal AT1R signaling, and reduce BP in SHR. SHR and wistar-kyoto (WKY) rats were treated with resveratrol for 8 weeks. Untreated SHR exhibited oxidative stress and enhanced renal proximal tubular Ang II-induced G-protein activation and Na/K-ATPase stimulation. Treatment of SHR with resveratrol mitigated oxidative stress, reduced BP, and normalized renal AT1R signaling. In SHR, nuclear expression of transcription factor NF-κB was increased while expression of Nrf2 was reduced. SHR also exhibited a significant decrease in renal antioxidant capacity and activities of phase II antioxidant enzymes. Resveratrol treatment of SHR abolished renal NF-κB activation, restored Nrf2-phase II antioxidant signaling and Ang II-mediated Na/K-ATPase regulation. These data show that in SHR, oxidative stress via activation of NF-κB upregulates AT1R-G-protein signaling resulting in overstimulation Na/K-ATPase which contributes to hypertension. Resveratrol, via Nrf2, activates phase II antioxidant enzymes, mitigates oxidative stress, normalizes AT1R-G-protein signaling and Na/K-ATPase regulation, and decreases BP in SHR.

7.
Am J Physiol Regul Integr Comp Physiol ; 308(10): R840-6, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25761698

RESUMO

Compelling evidence supports the role of oxidative stress and renal interstitial inflammation in the pathogenesis of hypertension. Resveratrol is a polyphenolic stilbene, which can lower oxidative stress by activating the transcription factor nuclear factor-E2-related factor-2 (Nrf2), the master regulator of numerous genes encoding antioxidant and phase II-detoxifying enzymes and molecules. Given the role of oxidative stress and inflammation in the pathogenesis of hypertension, we conducted this study to test the hypothesis that long-term administration of resveratrol will attenuate renal inflammation and oxidative stress and, hence, progression of hypertension in the young spontaneously hypertensive rats (SHR). SHR and control [Wistar-Kyoto (WKY)] rats were treated for 9 wk with resveratrol or vehicle in their drinking water. Vehicle-treated SHR exhibited renal inflammatory injury and oxidative stress, as evidenced by glomerulosclerosis, tubulointerstitial injury, infiltration of inflammatory cells, and increased levels of renal 8-isoprostane and protein carbonylation. This was associated with reduced antioxidant capacity and downregulations of Nrf2 and phase II antioxidant enzyme glutathione-S-transferase (GST). Resveratrol treatment mitigated renal inflammation and injury, reduced oxidative stress, normalized antioxidant capacity, restored Nrf2 and GST activity, and attenuated the progression of hypertension in SHR. However, resveratrol had no effect on these parameters in WKY rats. In conclusion, development and progression of hypertension in the SHR are associated with inflammation, oxidative stress, and impaired Nrf2-GST activity in the kidney. Long-term administration of resveratrol restores Nrf2 expression, ameliorates inflammation, and attenuates development of hypertension in SHR. Clinical studies are needed to explore efficacy of resveratrol in human hypertension.


Assuntos
Antioxidantes/uso terapêutico , Hipertensão/tratamento farmacológico , Inflamação/tratamento farmacológico , Rim/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Estilbenos/uso terapêutico , Animais , Antioxidantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Hipertensão/metabolismo , Inflamação/metabolismo , Rim/metabolismo , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Resveratrol , Estilbenos/farmacologia
8.
Hypertension ; 65(5): 1064-72, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25733244

RESUMO

There exists a strong link between oxidative stress, renal dopaminergic system, and hypertension. It is reported that reactive oxygen species attenuate renal proximal tubular dopamine receptor (D1R) function, which disrupts sodium regulation and leads to hypertension. However, the mechanisms for renal D1R dysfunction are not clear. We investigated the role of redox-sensitive transcription factors AP1 and SP3 in transcriptional suppression of D1R gene and subsequent D1R signaling. Human kidney proximal tubular cells were treated with a pro-oxidant l-buthionine sulfoximine (BSO) with and without an antioxidant tempol. In human kidney cells, BSO caused oxidative stress and reduced D1R mRNA and membrane receptor expression. Incubation of human kidney cells with SKF38393, a D1R agonist, caused a concentration-dependent inhibition of Na/K-ATPase. However, SKF38393 failed to inhibit Na/K-ATPase in BSO-treated cells. BSO increased AP1 and SP3 nuclear expression. Transfection with AP1- or SP3-specific siRNA abolished BSO-induced D1R downregulation. Treatment of rats with BSO for 4 weeks increased oxidative stress and SP3-AP1 expression and reduced D1R numbers in renal proximal tubules. These rats exhibited high blood pressure, and SKF38393 failed to inhibit proximal tubular Na/K-ATPase activity. Control rats were kept on tap water. Tempol per se had no effect on D1R expression or other signaling molecules but prevented BSO-induced oxidative stress, SP3-AP1 upregulation, and D1R dysfunction in both human kidney cells and rats. These data show that oxidative stress via AP1-SP3 activation suppresses D1R transcription and function. Tempol mitigates oxidative stress, blocks AP1-SP3 activation, and prevents D1R dysfunction and hypertension.


Assuntos
Hipertensão Renal/genética , Túbulos Renais Proximais/metabolismo , Estresse Oxidativo/genética , RNA Mensageiro/genética , Receptores de Dopamina D1/genética , Transcrição Gênica , Animais , Linhagem Celular , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Humanos , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Túbulos Renais Proximais/patologia , Masculino , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/biossíntese
9.
Hypertension ; 57(3): 452-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21282559

RESUMO

Oxidative stress modulates angiotensin (Ang) II type 1 receptor (AT(1)R) expression and function. Ang II activates renal Na(+)/H(+) exchanger 3 (NHE3) to increase sodium reabsorption, but the mechanisms are still elusive. In addition, the upregulation of AT(1)R during oxidative stress could promote sodium retention and lead to an increase in blood pressure. Herein, we investigated the mechanism of Ang II-mediated, AT(1)R-dependent renal NHE3 regulation and effect of oxidative stress on AT(1)R signaling and development of hypertension. Male Sprague-Dawley rats received tap water (control) or 30 mmol/L of l-buthionine-sulfoximine, an oxidant, with and without 1 mmol/L of Tempol, an antioxidant, for 3 weeks. l-Buthionine-sulfoximine-treated rats exhibited oxidative stress and high blood pressure. Incubation of renal proximal tubules with Ang II caused significantly higher NHE3 activation in l-buthionine-sulfoximine-treated rats compared with control. The activation of NHE3 was sensitive to AT(1)R blocker and inhibitors of phospholipase C, tyrosine kinase, janus kinase 2 (Jak2), Ca(2+)-dependent calmodulin (CaM), and Ca(2+) chelator. Also, incubation of proximal tubules with Ang II caused Jak2-dependent CaM phosphorylation, which led to Jak2-CaM complex formation and increased Jak2-CaM interaction with NHE3. The activation of these signaling molecules was exaggerated in l-buthionine-sulfoximine-treated rats, whereas Tempol normalized the AT(1)R signaling. In conclusion, Ang II activates renal proximal tubular NHE3 through novel pathways that involve phospholipase C and an increase in intracellular Ca(2+), Jak2, and CaM. In addition, oxidative stress exaggerates Ang II signaling, which leads to overstimulation of renal NHE3 and contributes to an increase in blood pressure.


Assuntos
Hipertensão/metabolismo , Rim/metabolismo , Estresse Oxidativo , Receptor Tipo 1 de Angiotensina/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Regulação para Cima , Análise de Variância , Animais , Pressão Sanguínea/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Janus Quinase 2/metabolismo , Rim/efeitos dos fármacos , Rim/fisiopatologia , Masculino , Malondialdeído/metabolismo , Fosforilação , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Trocador 3 de Sódio-Hidrogênio , Fosfolipases Tipo C/metabolismo
10.
Am J Physiol Renal Physiol ; 300(1): F98-104, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20926629

RESUMO

Impairment of renal dopamine D1 receptor (D1R)-mediated natriuresis is associated with hypertension in humans and animal models, including obese Zucker rats. We have previously reported that treatment of these rats with antioxidants or insulin sensitizers reduced insulin levels and oxidative stress, restored D1R-mediated natriuresis, and reduced blood pressure. Furthermore, the redox-sensitive transcription factor, nuclear factor-κB (NF-κB), has been implicated in impairment of D1R-mediated natriuresis during oxidative stress. In this study, we investigated the effect of exercise on insulin levels, oxidative stress, nuclear translocation of NF-κB, blood pressure, albuminuria, and D1R-mediated natriuresis. The exercise protocol involved treadmill exercise from 3 wk of age for 8 wk. Exercise reduced oxidative stress, nuclear translocation of NF-κB, and albuminuria. However, exercise did not reduce plasma insulin levels or blood pressure. Also, selective D1R agonist (SKF-38393)-mediated increases in sodium excretion and guanosine 5'-O-(3-thiotriphosphate) binding were impaired in obese rats compared with lean rats, and exercise did not restore this defect. We conclude that, while exercise is beneficial in reducing oxidative stress and renal injury, reducing insulin levels may be required to restore D1R-mediated natriuresis in this model of obesity and metabolic syndrome. Furthermore, this study supports previous observations that restoring D1R function contributes to blood pressure reduction in this model.


Assuntos
Hiperinsulinismo/prevenção & controle , Condicionamento Físico Animal/fisiologia , Receptores de Dopamina D1/fisiologia , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Albuminúria/prevenção & controle , Animais , Insulina/sangue , NF-kappa B/metabolismo , Estresse Oxidativo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Zucker , Receptores de Dopamina D1/agonistas
11.
Food Chem Toxicol ; 46(6): 2080-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18343012

RESUMO

Uranium, the heaviest of the naturally occurring elements is widely present as environmental contaminant from natural deposits, industrial emissions and most importantly from modern weapons. Histopathological examinations revealed that uranyl nitrate (UN) exposure caused severe damage to pars recta of renal proximal tubule. However, biochemical events involved in cellular response to renal injury are not completely elucidated. We hypothesized that UN exposure would severely damage kidney tissues and alter their metabolic functions. Rats were treated with a single nephrotoxic dose of UN (0.5mg/kg body weight) i.p. After 5d, effect of UN was studied on the activities of various enzymes of carbohydrate metabolism, brush border membrane (BBM) and oxidative stress in different kidney tissues. Activity of lactate dehydrogenase increased whereas activities of isocitrate, succinate and malate dehydrogenases, glucose-6-phosphatase and fructose-1,6-bisphosphatase significantly decreased by UN exposure. Activity of glucose-6-phosphate dehydrogenase decreased whereas that of NADP-malic enzyme increased. The activities of BBM enzymes were significantly lowered and after dissociation from BBM excreted in urine. Lipid peroxidation and the activities of superoxide dismutase and glutathione peroxidase increased whereas catalase activity decreased by UN. UN treatment caused specific alterations in the activities of metabolic and membrane enzymes and perturbed antioxidant defenses.


Assuntos
Metabolismo dos Carboidratos/efeitos dos fármacos , Rim/metabolismo , Microvilosidades/metabolismo , Nitrato de Uranil/farmacologia , Animais , Antioxidantes/metabolismo , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Eletrólitos/urina , Enzimas/sangue , Enzimas/urina , Glicosúria/induzido quimicamente , Rim/efeitos dos fármacos , Rim/enzimologia , Córtex Renal/efeitos dos fármacos , Córtex Renal/enzimologia , Córtex Renal/metabolismo , Masculino , Microvilosidades/efeitos dos fármacos , Microvilosidades/enzimologia , Ratos , Urodinâmica/efeitos dos fármacos
12.
Life Sci ; 82(9-10): 450-9, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18201728

RESUMO

Gentamicin (GM), an antibiotic against life threatening bacterial infection, induces remarkable toxicity in the kidney. Histological studies have indicated that mitochondria, microsomes, lysosomes and plasma membranes of renal proximal convoluted tubules in particular are major GM targets. Despite numerous investigations, the biochemical/cellular basis of GM nephrotoxicity is not well understood. Recently reactive oxygen species (ROS) are considered to be important mediators of GM-induced nephrotoxicity. We hypothesize that GM causes damage to intracellular organelles and affects their structural integrity and alters metabolic and other functional capabilities. To address above hypothesis a long-term, time-dependent effect of GM has been studied on blood/urine parameters, enzymes of carbohydrate metabolism, brush border membrane (BBM) and basolateral (BLM), lysosomes and oxidative stress in renal tissues. A nephrotoxic dose of GM (80 mg/kg body weight) was administered to rats daily for 15 days. The long-term treatment with GM induced a significant increase in serum creatinine, blood urea nitrogen followed by massive proteinuria, glucosuria, enzymuria along with loss of electrolytes in the urine. The activities of the enzymes of carbohydrate metabolism, plasma membranes, lysosomes significantly declined. The activities of antioxidant enzymes e.g. superoxide dismutase, catalase and glutathione peroxidase were severely depressed and lipid peroxidation was significantly increased in the renal cortex and medulla. We conclude that GM administration induced oxidative damage to renal tissues that resulted in impaired carbohydrate metabolism and decreased activities of BBM, BLM and lysosomes associated with increased lipid peroxides.


Assuntos
Gentamicinas/toxicidade , Rim/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Animais , Antibacterianos/administração & dosagem , Antibacterianos/toxicidade , Metabolismo dos Carboidratos/efeitos dos fármacos , Catalase/metabolismo , Gentamicinas/administração & dosagem , Glutationa Peroxidase/metabolismo , Injeções Intraperitoneais , Rim/metabolismo , Rim/patologia , Nefropatias/sangue , Nefropatias/induzido quimicamente , Nefropatias/urina , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Microvilosidades/efeitos dos fármacos , Microvilosidades/enzimologia , Microvilosidades/metabolismo , Potássio/sangue , Potássio/urina , Ratos , Ratos Wistar , Sódio/sangue , Sódio/urina , Superóxido Dismutase/metabolismo , Fatores de Tempo , gama-Glutamiltransferase/metabolismo
13.
Hypertension ; 51(2): 367-75, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18158345

RESUMO

Renal dopamine plays an important role in maintaining sodium homeostasis and blood pressure (BP) during increased sodium intake. The present study was carried out to determine whether renal dopamine D1 receptor (D1R) dysfunction contributes to increase in salt sensitivity during oxidative stress. Male Sprague-Dawley rats, divided into various groups, received tap water (vehicle); 1% NaCl (high salt [HS]); L-buthionine sulfoximine (BSO), an oxidant; and HS plus BSO with or without Tempol, an antioxidant, for 12 days. Compared with vehicle, HS intake increased urinary dopamine production and decreased basal renal Na/K-ATPase activity but did not affect BP. BSO-treated rats exhibited oxidative stress and a mild increase in BP. In these rats, D1R expression and G protein coupling were reduced, and SKF38393, a D1R agonist, failed to inhibit Na/K-ATPase activity and promote sodium excretion. Concomitant administration of BSO and HS caused oxidative stress, D1R dysfunction, and a marked increase in BP. Although renal dopamine production was increased, it failed to reduce the basal Na/K-ATPase activity in these animals. Treatment of BSO plus HS rats with Tempol decreased oxidative stress and restored endogenous, as well as exogenous, D1R agonist-mediated Na/K-ATPase inhibition and normalized BP. In conclusion, during HS intake, the increased dopamine production via Na/K-ATPase inhibition prevents an increase in BP. During oxidative stress, D1R function is defective, and there is mild hypertension. However, in the presence of oxidative stress, HS intake causes marked elevation in BP, which results from a defective renal D1R function leading to the failure of dopamine to inhibit Na/K-ATPase and promote sodium excretion.


Assuntos
Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Rim/metabolismo , Estresse Oxidativo , Receptores de Dopamina D1/metabolismo , Cloreto de Sódio , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Adenilil Ciclases/metabolismo , Animais , Antioxidantes/farmacologia , Butionina Sulfoximina/farmacologia , Óxidos N-Cíclicos/farmacologia , Diurese , Dopamina/urina , Agonistas de Dopamina/farmacologia , Ativação Enzimática , Proteínas de Ligação ao GTP/metabolismo , Hipertensão/fisiopatologia , Rim/enzimologia , Rim/fisiopatologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Masculino , Membranas/metabolismo , Natriurese , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/agonistas , ATPase Trocadora de Sódio-Potássio/metabolismo , Marcadores de Spin
14.
Biochim Biophys Acta ; 1716(1): 19-28, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16182235

RESUMO

The effect of ischemia induced acute renal failure (ARF) on the transport of phosphate (Pi) after early (15-30 min) and prolonged (60 min) ischemia in the brush border membrane vesicles (BBMV) from rat renal cortex was studied. Sodium-dependent transport of Pi declined significantly and progressively due to ischemia. Western blot analysis of BBM from ischemic rats showed decreased expression of NaPi-2. A compensatory increase was observed in Pi uptake in BBMV from contralateral kidneys. There was no significant difference in NaPi-2 expression between BBMV from sham and contralateral kidneys. Early blood reperfusion for 15 min after 30 min ischemia caused further decline in Pi uptake. Prolonged reperfusion for 120 min caused partial reversal of transport activities in 30-min ischemic rats. However, no improvement in the transport of Pi was observed in 60-min ischemic rats after 120 min of blood reperfusion. Kinetic studies showed that the effect of ischemia and blood reperfusion was dependent on the Vmax of the Na-Pi transporter. Western blot analysis showed increased expression of NaPi-2 in the BBMs from ischemia-reperfusion animals. Further, a shift in the association of Na ions to transport one molecule of Pi was observed under different extracellular Na concentrations [Na]o. Feeding rats with low Pi diet and/or treatment with thyroid hormone (T3) prior to ischemia resulted in increased basal Pi transport. Ischemia caused similar decline in Pi transport in BBM from LPD and/or T3 animals. However, recovery in these animals was faster than the normal Pi diet fed (NPD) animals. The study suggests a change in the intrinsic properties of the Na-Pi transporter in rat kidneys due to ischemia. The study also indicates that treatment with T3 and feeding LPD prior to ischemia caused faster recovery of phosphate uptake due to ischemia-reperfusion injury.


Assuntos
Membrana Celular/metabolismo , Rim/metabolismo , Fosfatos/química , Traumatismo por Reperfusão , Animais , Transporte Biológico , Western Blotting , Isquemia/patologia , Córtex Renal/metabolismo , Cinética , Microvilosidades , Fósforo/metabolismo , Prolina/química , Ratos , Ratos Wistar , Insuficiência Renal/metabolismo , Reperfusão , Sódio/química , Hormônios Tireóideos/metabolismo , Fatores de Tempo , Tri-Iodotironina/metabolismo
15.
Hypertension ; 40(6): 880-5, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12468573

RESUMO

Earlier we have reported a defective dopamine D1-like receptor function, which was accompanied by a decrease in D1 receptor numbers and the inability of dopamine to inhibit Na,K-ATPase and Na,H-exchanger in proximal tubules of hyperinsulinemic obese Zucker rats. The present study was designed to test the hypothesis that the defect in dopamine receptor function is a result of hyperinsulinemia in obese rats. We designed experiments to study D1 receptor function in obese Zucker rats treated with rosiglitazone, as it lowers plasma insulin by improving insulin sensitivity. A group of untreated lean and obese rats served as controls. Rosiglitazone treatment (10 mg/kg orally, 4 weeks) caused significant decreases in plasma insulin, blood glucose, and blood pressure while causing an increase in renal sodium excretion compared with untreated obese rats. In the isolated proximal tubules obtained from untreated lean rats, dopamine caused concentration-dependent inhibition of the Na,K-ATPase activity, but this inhibitory effect was absent in untreated obese rats. In rosiglitazone-treated obese rats, the inhibitory effect of dopamine on Na,K-ATPase was significantly restored. This was accompanied by a complete restoration of D1 receptor numbers in proximal tubular membranes of treated obese rats. In another set of experiments, treatment of primary proximal tubule epithelial cells in culture medium with insulin caused a significant decrease in the D1 receptor abundance, suggesting a direct role of insulin on D1 receptor regulation. We conclude that hyperinsulinemia causes downregulation of D1 receptor function and lowering of plasma insulin levels leads to restoration of renal D1 receptor function.


Assuntos
Rim/metabolismo , Obesidade/metabolismo , Receptores de Dopamina D1/metabolismo , Tiazóis/farmacologia , Tiazolidinedionas , Animais , Benzazepinas/farmacocinética , Ligação Competitiva/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Dopamina/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Glucose/farmacologia , Hiperinsulinismo/sangue , Hiperinsulinismo/tratamento farmacológico , Hipoglicemiantes/farmacologia , Insulina/sangue , Insulina/farmacologia , Rim/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Masculino , Ratos , Ratos Zucker , Rosiglitazona , ATPase Trocadora de Sódio-Potássio/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...