Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Hypertens ; 43(2): 175-180, 2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33070655

RESUMEN

Oxidative stress and renal inflammation play a pivotal role in the pathogenesis of hypertension. The redox-sensitive transcription factor, nuclear factor E2-related factor 2 (Nrf2) is the master regulator of phase II antioxidant enzymes that protects against oxidative stress and inflammation. This study aimed to investigate the effect of Nrf2 inhibition on oxidative stress-associated hypertension and renal dopamine 1 receptor (D1R) dysfunction in mice. Male C57BL/6 J mice were treated with a pro-oxidant, L-buthionine sulfoximine (BSO) (10 mmol/L in drinking water), and ML385 (10 kg body weight/kg body weight/day, intraperitoneally), a novel Nrf2 inhibitor that blocks Nrf2 regulated downstream target genes expression. Mice treated with BSO exhibited oxidative stress, renal functional impairment, inflammation, and elevated blood pressure. Also, BSO treatment increased the activity of phase II antioxidant enzyme, NAD(P)H: quinone oxidoreductase-1 (NQO-1). BSO and ML385 co-treatment exhibited a robust increase in blood pressure, oxidative stress and intensified the renal function deterioration as indicated by a significant increase in serum creatinine, urinary albumin excretion rate, and albumin to creatinine ratio and decreased glomerular filtration rate (GFR). Also, BSO and ML385 co-treatment downregulated NQO-1 and significantly altered the inflammatory cytokines, IL-1ß and IL-10 levels. A D1R agonist SKF38393 failed to promote urinary sodium excretion indicating functional impairment in renal D1R. ML385 per se did not affect mean arterial pressure, GFR, and renal D1R function. Taken together, we concluded that the Nrf2 inhibition aggravated oxidative stress and inflammation by diminishing phase II antioxidant defense that deteriorates renal function and contributes to the development of hypertension in mice.


Asunto(s)
Butionina Sulfoximina/farmacología , Hipertensión , Factor 2 Relacionado con NF-E2 , Especies Reactivas de Oxígeno , Animales , Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Inflamación/etiología , Inflamación/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/farmacología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología
2.
Am J Physiol Renal Physiol ; 319(4): F686-F696, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32830535

RESUMEN

Renal proximal tubular apoptosis plays a critical role in kidney health and disease. However, cellular molecules that trigger renal apoptosis remain elusive. Here, we evaluated the effect of inhibiting protein disulfide isomerase (PDI), a critical thioredoxin chaperone protein, on apoptosis as well as the underlying mechanisms in human renal proximal tubular (HK2) cells. HK2 cells were transfected with PDI-specific siRNA in the absence and presence of an antioxidant, tempol. PDI siRNA transfection resulted in a decrease of ~70% in PDI protein expression and enzyme activity. PDI inhibition increased caspase-3 activity and induced profound cell apoptosis. Mitochondrial function, as assessed by mitochondrial cytochrome c levels, mitochondrial membrane potential, oxygen consumption, and ATP levels, was significantly reduced in PDI-inhibited cells. Also, PDI inhibition caused nuclear factor erythroid 2-related factor 2 (Nrf2; a redox-sensitive transcription factor) cytoplasmic sequestration, decreased superoxide dismutase and glutathione-S-transferase activities, and increased oxidative stress. In PDI-inhibited cells, tempol reduced apoptosis, caspase-3 activity, and oxidative stress and also restored Nrf2 nuclear translocation and mitochondrial function. Silencing Nrf2 in the cells abrogated the beneficial effect of tempol, whereas Kelch-like ECH-associated protein 1 (an Nrf2 regulatory protein) silencing protected cells from PDI inhibitory effects. Collectively, our data indicate that PDI inhibition diminishes Nrf2 nuclear translocation, causing oxidative stress that further triggers mitochondrial dysfunction and renal cell apoptosis. This study suggests an important role for PDI in renal cell apoptosis involving Nrf2 and mitochondrial dysfunction.


Asunto(s)
Apoptosis , Células Epiteliales/enzimología , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Túbulos Renales Proximales/enzimología , Mitocondrias/enzimología , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Transporte Activo de Núcleo Celular , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Óxidos N-Cíclicos/farmacología , Metabolismo Energético , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/patología , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Proteína Disulfuro Isomerasas/genética , Interferencia de ARN , Transducción de Señal , Marcadores de Spin
3.
Clin Exp Hypertens ; 41(1): 5-13, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29424564

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Hipertensión/fisiopatología , Arterias Mesentéricas/fisiopatología , Proteínas de Microfilamentos/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo , Fosfoproteínas/metabolismo , Animales , Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Butionina Sulfoximina/farmacología , Calcio/metabolismo , Óxidos N-Cíclicos/farmacología , Masculino , Arterias Mesentéricas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/farmacología , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Marcadores de Spin , Canales Catiónicos TRPC/metabolismo , Vasodilatación/efectos de los fármacos
4.
Am J Physiol Regul Integr Comp Physiol ; 308(10): R840-6, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25761698

RESUMEN

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.


Asunto(s)
Antioxidantes/uso terapéutico , Hipertensión/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Riñón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Estilbenos/uso terapéutico , Animales , Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hipertensión/metabolismo , Inflamación/metabolismo , Riñón/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Resveratrol , Estilbenos/farmacología
5.
Clin Exp Hypertens ; 36(6): 367-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25198883

RESUMEN

Abstract The association of oxidative stress with hypertension is well known. However, a causal role of oxidative stress in hypertension is unclear. Vascular angiotensin II type 1 receptor (AT1R) upregulation is a prominent contributor to pathogenesis of hypertension. However, the mechanisms causing this upregulation are unknown. Oxidative stress is an important regulator of protein expression via activation of transcription factors such as nuclear factor kappa B (NFκB). The present study was carried out to test the hypothesis that oxidative stress contributes to vascular AT1R upregulation via NFκB in human aortic smooth muscle cells (HASMC) and spontaneously hypertensive rats (SHR). HASMC exposed to oxidative stress exhibited a robust increase in AT1R mRNA in HASMC. Furthermore, oxidative stress failed to upregulate AT1Rs in the presence of either an antioxidant catalase or siRNA against p65 subunit of NFκB. To test the role of oxidative stress and NFκB in hypertension, prehypertensive SHR were treated with NFκB inhibitor pyrrolidine dithiocarbamate from 5 weeks to 11-12 weeks of age. At 11-12 weeks of age, SHR exhibited increased NFκB expression, AT1R upregulation and exaggerated Ang II-induced vasoconstriction as compared to age-matched Wistar Kyoto (WKY) rats. PDTC treatment of SHR lowered NFκB expression, normalized AT1R expression and Ang II-induced vasoconstriction. More importantly, PDTC treatment significantly attenuated hypertension development in SHR. In conclusion, vascular oxidative can upregulate AT1R, via mechanisms involving NFκB, and contribute to the development of hypertension.


Asunto(s)
Hipertensión/fisiopatología , Músculo Liso Vascular/fisiopatología , FN-kappa B/fisiología , Estrés Oxidativo/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología , Animales , Aorta/efectos de los fármacos , Aorta/patología , Aorta/fisiopatología , Butionina Sulfoximina/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , FN-kappa B/efectos de los fármacos , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Prolina/análogos & derivados , Prolina/farmacología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tiocarbamatos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Vasoconstricción/fisiología
6.
Am J Physiol Renal Physiol ; 302(11): F1440-6, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22397926

RESUMEN

Nuclear factor-κB (NF-κB) plays a role in inflammation. However, we recently reported an association between NF-κB and antioxidant enzymes in renal proximal tubules of exercise-trained rats, suggesting its role in antioxidant homeostasis (George L, Lokhandwala MF, Asghar M. Am J Physiol Renal Physiol 297: F1174-F1180, 2009). A direct role of NF-κB in antioxidant homeostasis in renal cells has not been elucidated and warrants investigation. Therefore, we examined whether NF-κB has a direct role in antioxidant homeostasis and redox balance in human kidney-2 cells overexpressing NF-κB-p65 and compared them with the cells overexpressing Nrf-2, a well-known transcription factor involved in antioxidant homeostasis. The ability of NF-κB-p65 to increase antioxidant enzymes, to reduce reactive oxygen species (ROS), and to rescue ROS-induced renal dopamine D1 receptor dysfunction, was studied. The transcription activity of NF-κB-p65 and Nrf-2, measured as luciferase reporter activity, increased in cells overexpressing these nuclear factors. The levels of mRNA and activity of glutathione peroxidase as well as the protein levels of superoxide dismutase-1 and glutamylcystein transferase were increased in cells overexpressing NF-κB-p65 and Nrf-2. Furthermore, the levels of ROS decreased and D1 receptor agonist SKF38393-mediated [(35)S]GTPγS binding (index of D1 receptor function) increased in the presence of hydrogen peroxide in cells overexpressing NF-κB-p65 and Nrf-2. These results suggest a direct role of NF-κB-p65 in antioxidant homeostasis, contributing to redox balance in renal cells.


Asunto(s)
Antioxidantes/metabolismo , Homeostasis/fisiología , Riñón/metabolismo , Factor de Transcripción ReIA/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Western Blotting , Línea Celular , Agonistas de Dopamina/farmacología , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Glutatión Peroxidasa/biosíntesis , Glutatión Peroxidasa/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción ReIA/genética , Transfección
7.
Am J Physiol Renal Physiol ; 302(1): F47-51, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21900450

RESUMEN

Angiotensin (ANG) II via ANG II type 1 receptors (AT1R) activates renal sodium transporters including Na-K-ATPase and regulates sodium homeostasis and blood pressure. It is reported that at a high concentration, ANG II either inhibits or fails to stimulate Na-K-ATPase. However, the mechanisms for these phenomena are not clear. Here, we identified the signaling molecules involved in regulation of renal proximal tubular Na-K-ATPase at high ANG II concentrations. Proximal tubules from spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats were incubated with low concentrations of ANG II (pM), which activated Na-K-ATPase in both the groups; however, the stimulation was more robust in SHR. A high concentration of ANG II (µM) failed to stimulate Na-K-ATPase in WKY rats. However, in SHR ANG II (µM) continued to stimulate Na-K-ATPase, which was sensitive to the AT1R antagonist candesartan. In the presence of N(G)-nitro-l-arginine methyl ester (l-NAME), a nitric oxide (NO) synthase (NOS) inhibitor, ANG II (µM) caused stimulation of Na-K-ATPase in proximal tubules of WKY rats while having no further stimulatory effect in SHR. ANG II (µM), via AT1R, increased proximal tubular NO levels in WKY rats but not in SHR. In SHR, NOS was uncoupled as incubation of proximal tubules with ANG II and l-arginine, a NOS substrate, caused superoxide generation only in SHR and not in WKY rats. The superoxide production in SHR was sensitive to l-NAME. There was exaggerated proximal tubular AT1R-G protein coupling and NAD(P)H oxidase activation in response to ANG II (µM) in proximal tubules of SHR compared with WKY rats. In SHR, inhibition of NADPH oxidase restored NOS coupling and ANG II-induced NO accumulation. In conclusion, at a high concentration ANG II (µM) activates renal NO signaling, which prevents stimulation of Na-K-ATPase in WKY rats. However, in SHR ANG II (µM) overstimulates NADPH oxidase, which impairs the NO system and leads to continued Na-K-ATPase activation.


Asunto(s)
Angiotensina II/fisiología , Túbulos Renales Proximales/fisiopatología , Óxido Nítrico/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Arginina/metabolismo , Bencimidazoles/farmacología , Compuestos de Bifenilo , Túbulos Renales Proximales/efectos de los fármacos , Masculino , NADPH Oxidasas/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/metabolismo , Tetrazoles/farmacología
8.
Antioxidants (Basel) ; 11(1)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35052660

RESUMEN

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.

9.
Am J Physiol Renal Physiol ; 300(1): F133-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20943769

RESUMEN

Aging is associated with an increase in oxidative stress and blood pressure (BP). Renal dopamine D1 (D1R) and angiotensin II AT1 (AT1R) receptors maintain sodium homeostasis and BP. We hypothesized that age-associated increase in oxidative stress causes altered D1R and AT1R functions and high BP in aging. To test this, adult (3 mo) and old (21 mo) Fischer 344 × Brown Norway F1 rats were supplemented without/with antioxidant tempol followed by determining oxidative stress markers (urinary antioxidant capacity, proximal tubular NADPH-gp91phox, and plasma 8-isoprostane), D1R and AT1R functions, and BP. The D1R and AT1R functions were determined by measuring diuretic and natriuretic responses to D1R agonist (SKF-38393; 1 µg·kg(-1)·min(-1) iv) and AT1R antagonist (candesartan; 10 µg/kg iv), respectively. We found that the total urinary antioxidant capacity was lower in old rats, which increased with tempol treatment. In addition, tempol decreased the elevated NADPH-gp91phox and 8-isoprostane levels in old rats. Systolic, diastolic, and mean arterial BPs were higher in old rats and were reduced by tempol. Although SKF-38393 produced diuresis in both adult and old rats, urinary sodium excretion (UNaV) increased only in adult rats. While candesartan increased diuresis and UNaV in adult and old rats, the magnitude of response was greater in old rats. Tempol treatment in old rats reduced candesartan-induced increase in diuresis and UNaV. Our results demonstrate that diminished renal D1R and exaggerated AT1R functions are associated with high BP in old rats. Furthermore, oxidative stress may cause altered renal D1R and AT1R functions and high BP in old rats.


Asunto(s)
Envejecimiento/fisiología , Presión Sanguínea/efectos de los fármacos , Estrés Oxidativo/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Receptores de Dopamina D1/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Bencimidazoles/farmacología , Compuestos de Bifenilo , Óxidos N-Cíclicos/farmacología , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Frecuencia Cardíaca/fisiología , Túbulos Renales Proximales/fisiología , Masculino , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/efectos de los fármacos , NADPH Oxidasas/metabolismo , Natriuresis/efectos de los fármacos , Ratas , Receptores de Dopamina D1/agonistas , Marcadores de Spin , Tetrazoles/farmacología
10.
Am J Physiol Renal Physiol ; 301(2): F364-70, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21593187

RESUMEN

Angiotensin (ANG) II via AT1 receptors (AT1Rs) maintains sodium homeostasis by regulating renal sodium transporters including Na(+)/H(+) exchanger 3 (NHE3) in a biphasic manner. Low-ANG II concentration stimulates whereas high concentrations inhibit NHE3 activity. Oxidative stress has been shown to upregulate AT1R function that could modulate the ANG II-mediated NHE3 regulation. This study was designed to identify the signaling pathways responsible for ANG II-mediated biphasic regulation of proximal tubular NHE3 and the effect of oxidative stress on this phenomenon. Male Sprague-Dawley rats were chronically treated with a pro-oxidant L-buthionine sulfoximine (BSO) with and without an antioxidant tempol in tap water for 3 wk. BSO-treated rats exhibited oxidative stress and high blood pressure. At low concentration (1 pM) ANG II increased NHE3 activity in proximal tubules from all animals. However, in BSO-treated rats, the stimulation was more robust and was normalized by tempol treatment. ANG II (1 pM)-mediated NHE3 activation was abolished by AT1R blocker, intracellular Ca(2+) chelator, and inhibitors of phospholipase C (PLC) and Ca(2+)-dependent calmodulin (CaM) but it was insensitive to Giα and protein kinase C inhibitors or AT2R antagonist. A high concentration of ANG II (1 µM) inhibited NHE3 activity in control and tempol-treated rats. However, in BSO-treated rats, ANG II (1 µM) continued to induce NHE3 stimulation. Tempol restored the inhibitory effect of ANG II (1 µM) in BSO-treated rats. The inhibitory effect of ANG II (1 µM) involved AT1R-dependent, cGMP-dependent protein kinase (PKG) activation and was independent of AT2 receptor and nitric oxide signaling. We conclude that ANG II stimulates NHE3 via AT1R-PLC-CaM pathway and inhibits NHE3 by AT1R-PKG activation. Oxidative stress impaired ANG II-mediated NHE3 biphasic response in that stimulation was observed at both high- and low-ANG II concentration.


Asunto(s)
Angiotensina II/metabolismo , Túbulos Renales Proximales/metabolismo , Estrés Oxidativo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activación Enzimática , Masculino , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Intercambiador 3 de Sodio-Hidrógeno
11.
Am J Physiol Renal Physiol ; 300(1): F98-104, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20926629

RESUMEN

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.


Asunto(s)
Hiperinsulinismo/prevención & control , Condicionamiento Físico Animal/fisiología , Receptores de Dopamina D1/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Albuminuria/prevención & control , Animales , Insulina/sangre , FN-kappa B/metabolismo , Estrés Oxidativo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Zucker , Receptores de Dopamina D1/agonistas
12.
Curr Hypertens Rep ; 13(4): 294-302, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21633929

RESUMEN

The role of dopamine receptors in blood pressure regulation is well established. Genetic ablation of both dopamine D1-like receptor subtypes (D1, D5) and D2-like receptor subtypes (D2, D3, D4) results in a hypertensive phenotype in mice. This review focuses on the dopamine D1-like receptor subtypes D1 and D5 (especially D1 receptors), as they play a major role in regulating sodium homeostasis and blood pressure. Studies mostly describing the role of renal dopamine D1-like receptors are included, as the kidneys play a pivotal role in the maintenance of sodium homeostasis and the long-term regulation of blood pressure. We also attempt to describe the interaction between D1-like receptors and other proteins, especially angiotensin II type 1 and type 2 receptors, which are involved in the maintenance of sodium homeostasis and blood pressure. Finally, we discuss a new concept of renal D1 receptor regulation in hypertension that involves oxidative stress mechanisms.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Receptores de Dopamina D1/biosíntesis , Quinasas de Receptores Acoplados a Proteína-G , Homeostasis , Humanos , Hipertensión/genética , Estrés Oxidativo , Receptor de Angiotensina Tipo 1 , Receptor de Angiotensina Tipo 2 , Receptores de Dopamina D1/efectos de los fármacos
13.
J Am Heart Assoc ; 9(1): e014977, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31880979

RESUMEN

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.


Asunto(s)
Presión Sanguínea , Dopamina/metabolismo , Hipertensión/etiología , Túbulos Renales Proximales/metabolismo , Nefritis/etiología , Estrés Oxidativo , Cloruro de Sodio Dietético/toxicidad , Animales , Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/prevención & control , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Lisosomas/metabolismo , Masculino , Mitocondrias/metabolismo , Nefritis/metabolismo , Nefritis/fisiopatología , Nefritis/prevención & control , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley
14.
Am J Physiol Renal Physiol ; 297(6): F1543-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19794106

RESUMEN

We tested the effects of inflammation on renal dopamine D1 receptor signaling cascade, a key pathway that maintains sodium homeostasis and blood pressure during increased salt intake. Inflammation was produced by administering lipopolysaccharide (LPS; 4 mg/kg ip) to rats provided without (normal salt) and with 1% NaCl in drinking water for 2 wk (high salt). Control rats had saline injection and received tap water. We found that LPS increased the levels of inflammatory cytokines, interleukin-6, and tumor necrosis factor-alpha in the rats given either normal- or high-salt intake. Also, these rats had higher levels of oxidative stress markers, malondialdehyde and nitrotyrosine, and lower levels of antioxidant enzyme superoxide dismutase in the renal proximal tubules (RPTs). The nuclear levels of transcription factors NF-kappaB increased and Nrf2 decreased in the RPTs in response to LPS in rats given normal and high salt. Furthermore, D1 receptor numbers, D1 receptor proteins, and D1 receptor agonist (SKF38393)-mediated (35)S-GTPgammaS binding decreased in the RPTs in these rats. The basal activities of Na-K-ATPase in the RPTs were similar in control and LPS-treated rats given normal and high salt. SKF38393 caused inhibition of Na-K-ATPase activity in the primary cultures of RPTs treated with vehicle but not in the cultures treated with LPS. Furthermore, LPS caused an increase in blood pressure in the rats given high salt but not in the rats given normal salt. These results suggest that LPS differentially regulates NF-kappaB and Nrf2, produces inflammation, decreases antioxidant enzyme, increases oxidative stress, and causes D1 receptor dysfunction in the RPTs. The LPS-induced dysfunction of renal D1 receptors alters salt handling and causes hypertension in rats during salt overload.


Asunto(s)
Inflamación/metabolismo , Riñón/metabolismo , Receptores de Dopamina D1/metabolismo , Transducción de Señal , Animales , Hipertensión/inducido químicamente , Inflamación/inducido químicamente , Túbulos Renales Proximales/metabolismo , Lipopolisacáridos/farmacología , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Cloruro de Sodio/metabolismo
15.
Am J Physiol Renal Physiol ; 297(5): F1174-80, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19759268

RESUMEN

We have previously reported that age-associated oxidative stress via protein kinase C (PKC) increases D1 receptor (D1R) phosphorylation and causes D1R-G protein uncoupling in renal proximal tubules (RPTs) of old Fischer 344 rats. This results in reduced ability of D1R agonist SKF-38393 to inhibit Na+-K+-ATPase in RPTs of old rats. Here, we studied the effect of treadmill exercise on markers of oxidative stress, PKC, D1R phosphorylation, D1R-G protein coupling, and Na+-K+-ATPase activity in RPTs of adult and old rats. We found increased levels of malondialdehyde, a marker of oxidative stress, in RPTs of old rats, which decreased during exercise. Nuclear levels of nuclear erythroid-related factor (Nrf)-2 and nuclear factor (NF)-kappaB in RPTs, transcription factors involved in antioxidant enzyme gene transcription, increased in exercised old rats. This was accompanied by an increase in the activity and expression of antioxidant enzymes, superoxide dismutase and heme oxygenase-1. Age-related decrease in the levels of D1R mRNAs and proteins was attenuated during exercise. Furthermore, exercise in old rats decreased PKC activity and D1R phosphorylation and increased SKF-38393-mediated [35S]guanosine 5'-O-(3-thiotriphosphate) binding (an index of D1R-G protein coupling). SKF-38393 also caused inhibition of Na+-K+-ATPase in these animals. Also, exercise caused a decrease in proteinuria and increase in phosphaturia in old rats. These results suggest beneficial effects of exercise in terms of increasing antioxidant defenses, decreasing oxidative stress, and improving kidney function in general and D1R function in particular in aging. Both Nrf-2 and NF-kappaB seem to play key role in this phenomenon.


Asunto(s)
Envejecimiento , Condicionamiento Físico Animal , Receptores de Dopamina D1 , Factores de Transcripción , Animales , Masculino , Ratas , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Envejecimiento/fisiología , Agonistas de Dopamina/farmacología , Proteínas de Unión al GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/metabolismo , Malondialdehído/metabolismo , FN-kappa B/metabolismo , Oxidación-Reducción , Fosfatos/orina , Condicionamiento Físico Animal/fisiología , Proteína Quinasa C/metabolismo , Proteinuria/metabolismo , Ratas Endogámicas F344 , Receptores de Dopamina D1/fisiología , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo , Factores de Transcripción/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo
16.
Am J Physiol Renal Physiol ; 297(2): F397-402, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19474184

RESUMEN

Renal dopamine and nitric oxide contribute to natriuresis during high-salt intake which maintains sodium and blood pressure homeostasis. We wanted to determine whether concurrent inhibition of these natriuretic factors increases blood pressure during high-sodium intake. Male Sprague-Dawley rats were divided into the following groups: 1) vehicle (V)-tap water, 2) NaCl-1% NaCl drinking water, 3) 30 mM l-buthionine sulfoximine (BSO), an oxidant, 4) BSO plus NaCl, and 5) BSO plus NaCl with 1 mM tempol (antioxidant). Compared with V, NaCl intake for 10 days doubled sodium intake and increased urinary dopamine level but reduced urinary nitric oxide content. NaCl intake also reduced basal renal proximal tubular Na-K-ATPase activity with no effect on blood pressure. However, NaCl intake in BSO-treated rats failed to reduce basal Na-K-ATPase activity despite higher urinary dopamine levels. Also, dopamine failed to inhibit proximal tubular Na-K-ATPase activity and these rats exhibited reduced urinary nitric oxide levels and high blood pressure. Tempol supplementation in NaCl plus BSO-treated rats reduced blood pressure. BSO treatment alone did not affect the urinary nitric oxide and dopamine levels or blood pressure. However, dopamine failed to inhibit proximal tubular Na-K-ATPase activity in BSO-treated rats. BSO treatment also increased basal protein kinase C activity, D1 receptor serine phosphorylation, and oxidative markers like malondialdehyde and 8-isoprostane. We suggest that NaCl-mediated reduction in nitric oxide does not increase blood pressure due to activation of D1 receptor signaling. Conversely, oxidative stress-provoked inhibition of D1 receptor signaling fails to elevate blood pressure due to presence of normal nitric oxide. However, simultaneously decreasing nitric oxide levels with NaCl and inhibiting D1 receptor signaling with BSO elevated blood pressure.


Asunto(s)
Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Dopamina/orina , Hipertensión/fisiopatología , Natriuresis/efectos de los fármacos , Óxido Nítrico/orina , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Amidohidrolasas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/orina , Butionina Sulfoximina/farmacología , Óxidos N-Cíclicos/farmacología , Modelos Animales de Enfermedad , Hipertensión/etiología , Hipertensión/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/fisiopatología , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Serina , Transducción de Señal/efectos de los fármacos , Cloruro de Sodio Dietético/administración & dosificación , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Marcadores de Spin
17.
Curr Hypertens Rep ; 10(4): 268-75, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18625155

RESUMEN

Dopamine plays an important role in regulating renal function and blood pressure. Dopamine synthesis and dopamine receptor subtypes have been shown in the kidney. Dopamine acts via cell surface receptors coupled to G proteins; the receptors are classified via pharmacologic and molecular cloning studies into two families, D1-like and D2-like. Two D1-like receptors cloned in mammals, the D1 and D5 receptors (D1A and D1B in rodents), are linked to adenylyl cyclase stimulation. Three D2-like receptors (D2, D3, and D4) have been cloned and are linked mainly to adenylyl cyclase inhibition. Activation of D1-like receptors on the proximal tubules inhibits tubular sodium reabsorption by inhibiting Na/H-exchanger and Na/K-adenosine triphosphatase activity. Reports exist of defective renal dopamine production and/or dopamine receptor function in human primary hypertension and in genetic models of animal hypertension. In humans with essential hypertension, renal dopamine production in response to sodium loading is often impaired and may contribute to hypertension. A primary defect in D1-like receptors and an altered signaling system in proximal tubules may reduce dopamine-mediated effects on renal sodium excretion. The molecular basis for dopamine receptor dysfunction in hypertension is being investigated, and may involve an abnormal posttranslational modification of the dopamine receptor.


Asunto(s)
Hipertensión/fisiopatología , Receptores Dopaminérgicos/fisiología , Animales , Cardiotónicos/farmacología , Dopamina/farmacología , Dopamina/fisiología , Dopamina/orina , Humanos , Riñón/efectos de los fármacos , Riñón/fisiopatología , Natriuresis/fisiología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología
18.
Diabetes ; 54(7): 2219-26, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15983225

RESUMEN

Oxidative stress plays a pathogenic role in hypertension, particularly the one associated with diabetes and obesity. Here, we test the hypothesis that renal dopamine D1 receptor dysfunction in obese Zucker rats is caused by oxidative stress. One group each from lean and obese Zucker rats received tempol, a superoxide dismutase mimetic in drinking water for 2 weeks. Obese animals were hypertensive, hyperglycemic, and hyperinsulinemic, exhibited renal oxidative stress, and increased protein kinase C activity. Also, there was hyperphosphorylation of D1 receptor, defective receptor-G-protein coupling, blunted dopamine-induced Na+-K+-ATPase inhibition, and diminished natriuretic response to D1 receptor agonist, SKF-38393. However, obese animals had elevated levels of plasma nitric oxide and urinary cGMP. In addition, L-N-nitroarginine and sodium nitroprusside showed similar effect on blood pressure in lean and obese rats. In obese animals, tempol reduced blood pressure, blood glucose, insulin, renal oxidative stress, and protein kinase C activity. Tempol also decreased D1 receptor phosphorylation and restored receptor G-protein coupling. Dopamine inhibited Na+-K+-ATPase activity, and SKF-38393 elicited a natriuretic response in tempol-treated obese rats. Thus in obese Zucker rats, tempol ameliorates oxidative stress and improves insulin sensitivity. Consequently, hyperphosphorylation of D1 receptor is reduced, leading to restoration of receptor-G-protein coupling and the natriuretic response to SKF-38393.


Asunto(s)
Óxidos N-Cíclicos/farmacología , Proteínas de Unión al GTP/metabolismo , Insulina/farmacología , Riñón/fisiología , Obesidad/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Receptores de Dopamina D1/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Insulina/sangre , Riñón/efectos de los fármacos , Masculino , Malondialdehído/sangre , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Zucker , Receptores de Dopamina D1/efectos de los fármacos , Marcadores de Spin
19.
Free Radic Biol Med ; 40(1): 13-20, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16337875

RESUMEN

Dopamine, via activation of D1-like receptors, inhibits Na,K-ATPase and Na,H-exchanger in renal proximal tubules and promotes sodium excretion. This effect of dopamine is not seen in conditions associated with oxidative stress such as hypertension, diabetes, and aging due to uncoupling of D1-like receptors from G proteins. To identify the role of oxidative stress in uncoupling of the D1-like receptors, we utilized primary cultures from rat renal proximal tubules. Hydrogen peroxide (H2O2), an oxidant, treatment to the cell cultures increased the level of malondialdehyde, a marker of oxidative damage. Further, H2O2 decreased membranous D1-like receptor numbers and proteins, D1-like agonist (SKF 38393)-mediated [35S]GTPgammaS binding and SKF 38393-mediated inhibition of Na,K-ATPase. Moreover, H2O2 treatment to the cultures caused membranous translocation of G-protein-coupled receptor kinase 2 (GRK 2) and increased serine phosphorylation of D1A receptors accompanied by an increase in protein kinase C (PKC) activity. Interestingly, PKC inhibitors blocked the H2O2-mediated stimulation of GRK 2 and serine phosphorylation of D1A receptors. Further, GRK 2 antisense but not scrambled oligonucleotides attenuated the effect of H2O2 on membranous expression of GRK 2. Moreover, direct activation of PKC with phorbol ester (PMA) resulted in reduction of SKF 38393-mediated [35S]GTPgammaS binding. We conclude that H2O2 stimulates PKC leading to the activation of GRK 2, which causes serine phopshorylation of D1A receptors and receptor G-protein uncoupling in these cells, resulting in impairment in D1-like receptor function.


Asunto(s)
Peróxido de Hidrógeno/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Oxidantes/farmacología , Proteína Quinasa C/metabolismo , Receptores de Dopamina D1/metabolismo , Quinasas de Receptores Adrenérgicos beta/metabolismo , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Animales , Agonistas de Dopamina/farmacología , Inhibidores Enzimáticos/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Túbulos Renales Proximales/metabolismo , Malondialdehído/metabolismo , Estrés Oxidativo , Ésteres del Forbol/farmacología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Quinasas de Receptores Adrenérgicos beta/antagonistas & inhibidores
20.
Hypertension ; 65(5): 1064-72, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25733244

RESUMEN

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.


Asunto(s)
Hipertensión Renal/genética , Túbulos Renales Proximales/metabolismo , Estrés Oxidativo/genética , ARN Mensajero/genética , Receptores de Dopamina D1/genética , Transcripción Genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Humanos , Hipertensión Renal/metabolismo , Hipertensión Renal/patología , Túbulos Renales Proximales/patología , Masculino , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA