Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Curr Opin Nephrol Hypertens ; 20(5): 529-33, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21670672

RESUMO

PURPOSE OF REVIEW: Activation of epithelial sodium channel (ENaC) by proteolysis appears to be relevant for day-to-day physiological regulation of channel activity in kidney and other epithelial tissues. Pathophysiogical, proteolytic activation of ENaC in kidney has been demonstrated in proteinuric disease. RECENT FINDINGS: A variation in sodium and potassium intake or plasma aldosterone changes the number of cleaved α and γ-ENaC subunits and is associated with changes in ENaC currents. The protease furin mediates intracellular cleavage, whereas the channel-activating protease prostasin (CAP-1), which is glycophosphatidylinositol-anchored to the apical cell surface, mediates important extracellular cleavage. Soluble protease activity is very low in urine under physiological conditions but rises in proteinuria. In nephrotic syndrome, the dominant soluble protease activity is plasmin, which is formed from filtered plasminogen via urokinase-type plasminogen activator. Plasmin activates ENaC directly at high concentrations and through prostasin at lower concentrations. SUMMARY: The discovery of serine protease-mediated activation of renal ENaC in physiological and pathophysiological conditions opens the way for new understanding of the pathogenesis of proteinuric sodium retention, which may involve plasmin and present several potential new drug targets.


Assuntos
Células Epiteliais/enzimologia , Canais Epiteliais de Sódio/metabolismo , Ativação do Canal Iônico , Túbulos Renais/enzimologia , Peptídeo Hidrolases/metabolismo , Aldosterona/metabolismo , Animais , Humanos , Transporte de Íons , Túbulos Renais/fisiopatologia , Potássio/metabolismo , Proteinúria/enzimologia , Proteinúria/fisiopatologia , Sódio/metabolismo
2.
Am J Hypertens ; 23(11): 1241-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20651700

RESUMO

BACKGROUND: Mice with targeted deletion of neuronal nitric oxide (NO) synthase (nNOS⁻(/)⁻) display inability to increase plasma renin concentration (PRC) in response to sodium restriction. nNOS has a distinct expression at the macula densa (MD), and in the present study, it was tested whether nNOS supports renin release by cyclic guanosine monophosphate (cGMP)-mediated inhibition of cyclic adenosine monophosphate (cAMP)-specific phosphodiesterase 3 (PDE3) in juxtaglomerular (JG) cells. METHODS: The experiments were performed in conscious nNOS⁻(/)⁻ and wild types after 10 days on a low-sodium diet by acute treatment with the PDE3-inhibitor milrinone, the PDE5 inhibitor zaprinast, or vehicle, using a crossover study protocol. PRC was measured with the antibody-trapping technique and blood pressure with telemetry. Glomerular filtration rate (GFR) and renal plasma flow (RPF) were estimated by measurements of inulin- and para-amino hippuric acid (PAH) clearances, respectively. RESULTS: The basal PRC was reduced in nNOS⁻(/)⁻ compared to the wild types. Administration of milrinone caused a more pronounced PRC increase in nNOS⁻(/)⁻, resulting in normalized renin levels, whereas PDE5 inhibition did not affect PRC in any genotype. The blood pressure was similar in both genotypes, and milrinone did not affect blood pressure compared to vehicle. GFR and RPF were similar at baseline and were reduced by milrinone. CONCLUSIONS: The present study provides in vivo evidence supporting the view that NO, selectively derived from nNOS, mediates renin release during sodium restriction by inhibiting PDE3, which would increase renin release by elevating cAMP levels in the JG cells.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Renina/metabolismo , Cloreto de Sódio na Dieta/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Feminino , Sistema Justaglomerular/fisiologia , Córtex Renal/efeitos dos fármacos , Córtex Renal/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Milrinona/farmacologia , Inibidores da Fosfodiesterase 3/farmacologia , Inibidores da Fosfodiesterase 5/farmacologia , Purinonas/farmacologia , Circulação Renal/efeitos dos fármacos , Circulação Renal/fisiologia
3.
J Hepatol ; 52(5): 705-11, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20347173

RESUMO

BACKGROUND & AIMS: Suppression of the hypothalamic-pituitary-adrenal axis occurs in cirrhosis and cholestasis and is associated with increased concentrations of bile acids. We investigated whether this was mediated through bile acids acting to impair steroid clearance by inhibiting glucocorticoid metabolism by 5beta-reductase. METHODS: The effect of bile acids on glucocorticoid metabolism was studied in vitro in hepatic subcellular fractions and hepatoma cells, allowing quantitation of the kinetics and transcript abundance of 5beta-reductase. Metabolism was subsequently examined in vivo in rats following dietary manipulation or bile duct ligation. Finally, glucocorticoid metabolism was assessed in humans with obstructive jaundice. RESULTS: In rat hepatic cytosol, chenodeoxycholic acid competitively inhibited 5beta-reductase (K(i) 9.19+/-0.40 microM) and reduced its transcript abundance (in H4iiE cells) and promoter activity (reporter system, HepG2 cells). In Wistar rats, dietary chenodeoxycholic acid (1% w/w chow) inhibited hepatic 5beta-reductase activity, reduced urinary excretion of 3alpha,5beta-tetrahydrocorticosterone and reduced adrenal weight. Conversely, a fat-free diet suppressed bile acid levels and increased hepatic 5beta-reductase activity, supplementation of the fat-free diet with CDCA reduced 5beta-reductase activity, and urinary 3alpha,5beta-reduced corticosterone. Cholestasis in rats suppressed hepatic 5beta-reductase activity and transcript abundance. In eight women with obstructive jaundice, relative urinary excretion of 3alpha,5beta-tetrahydrocortisol was significantly lower than in healthy controls. CONCLUSION: These data suggest a novel role for bile acids in inhibiting hepatic glucocorticoid clearance, of sufficient magnitude to suppress hypothalamic-pituitary-adrenal axis activity. Elevated hepatic bile acids may account for adrenal insufficiency in liver disease.


Assuntos
Ácidos e Sais Biliares/farmacologia , Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Icterícia Obstrutiva/tratamento farmacológico , Sistema Hipófise-Suprarrenal/fisiologia , 3-Hidroxiesteroide Desidrogenases/genética , Animais , Sequência de Bases , Ácidos e Sais Biliares/uso terapêutico , Ductos Biliares/fisiologia , Citosol/enzimologia , Feminino , Humanos , Hidrocortisona/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Icterícia Obstrutiva/metabolismo , Icterícia Obstrutiva/urina , Cinética , Ligadura , Fígado/enzimologia , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ratos , Ratos Wistar , Tetra-Hidrocortisol/urina , Transcrição Gênica/efeitos dos fármacos
4.
Kidney Int ; 77(2): 110-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19907416

RESUMO

To examine the role of the calcium/calmodulin-dependent phosphatase calcineurin in regulation of renin release, we assayed exocytosis using whole-cell patch clamp of single juxtaglomerular cells in culture. The calcineurin inhibitor, cyclosporine A (CsA), significantly increased juxtaglomerular cell membrane capacitance, an index of cell surface area and an established measure of exocytosis in single-cell assays. This effect was mimicked by intracellular delivery of a calcineurin inhibitory peptide, the calcium chelator ethylene glycol tetraacetic acid (EGTA), or the calmodulin inhibitor W-13. Simultaneous exposure to EGTA and CsA had no additive effect. The protein kinase A (PKA) blocker RpcAMPs had no effect on the CsA-induced increase in membrane capacitance. Intra- and extracellular application of tacrolimus did not alter membrane capacitance. A calmodulin antagonist (calmidazolium) and CsA, but not tacrolimus, significantly stimulated renin release from cultured juxtaglomerular cells. Juxtaglomerular cells expressed the calcineurin isoforms A-beta and A-gamma but not A-alpha. Plasma renin concentrations (PRCs) were not different in wild-type, calcineurin A-alpha, or A-beta knockout mice but increased after CsA treatment of the A-alpha knockout, while renin mRNA was suppressed. We conclude that calcineurin and calcium/calmodulin suppress exocytosis of renin from juxtaglomerular cells independent of PKA.


Assuntos
Calcineurina/metabolismo , Cálcio/metabolismo , Exocitose , Sistema Justaglomerular/metabolismo , Renina/metabolismo , Animais , Calcineurina/genética , Cálcio/antagonistas & inibidores , Calmodulina/antagonistas & inibidores , Células Cultivadas , Quelantes , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Ciclosporina , Ácido Egtázico , Capacitância Elétrica , Imunossupressores , Masculino , Camundongos , Técnicas de Patch-Clamp , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Renina/sangue , Sulfonamidas
5.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1733-41, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19793956

RESUMO

Several pathophysiological conditions, including nephrotic syndrome, are characterized by increased renal activity of the epithelial Na(+) channel (ENaC). We recently identified plasmin in nephrotic urine as a stimulator of ENaC activity and undertook this study to investigate the mechanism by which plasmin stimulates ENaC activity. Cy3-labeled plasmin was found to bind to the surface of the mouse cortical collecting duct cell line, M-1. Binding depended on a glycosylphosphatidylinositol (GPI)-anchored protein. Biotin-label transfer showed that plasmin interacted with the GPI-anchored protein prostasin on M-1 cells and that plasmin cleaved prostasin. Prostasin activates ENaC by cleavage of the gamma-subunit, which releases an inhibitory peptide from the extracellular domain. Removal of GPI-anchored proteins from the M-1 cells with phosphatidylinositol-specific phospholipase C (PI-PLC) inhibited plasmin-stimulated ENaC current in monolayers of M-1 cells at low plasmin concentration (1-4 microg/ml). At a high plasmin concentration of 30 microg/ml, there was no difference between cell layers treated with or without PI-PLC. Knockdown of prostasin attenuated binding of plasmin to M1 cells and blocked plasmin-stimulated ENaC current in single M-1 cells, as measured by whole-cell patch clamp. In M-1 cells expressing heterologous FLAG-tagged prostasin, gammaENaC and prostasin were colocalized. A monoclonal antibody directed against the inhibitory peptide of gammaENaC produced specific immunofluorescence labeling of M-1 cells. Pretreatment with plasmin abolished labeling of M-1 cells in a prostasin-dependent way. We conclude that, at low concentrations, plasmin interacts with GPI-anchored prostasin, which leads to cleavage of the gamma-subunit and activation of ENaC, while at higher concentrations, plasmin directly activates ENaC.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Fibrinolisina/metabolismo , Ativação do Canal Iônico , Túbulos Renais Coletores/metabolismo , Serina Endopeptidases/metabolismo , Animais , Biotinilação , Carbocianinas/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Imunofluorescência , Corantes Fluorescentes/metabolismo , Túbulos Renais Coletores/citologia , Potenciais da Membrana , Camundongos , Síndrome Nefrótica/urina , Oligopeptídeos , Técnicas de Patch-Clamp , Peptídeos/metabolismo , Fosfoinositídeo Fosfolipase C/metabolismo , Ligação Proteica , Interferência de RNA , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/genética , Fatores de Tempo
6.
J Am Soc Nephrol ; 20(10): 2154-61, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628672

RESUMO

The mechanism by which extracellular hypotonicity stimulates release of renin from juxtaglomerular (JG) cells is unknown. We hypothesized that osmotically induced renin release depends on water movement through aquaporin-1 (AQP1) water channels and subsequent prostanoid formation. We recorded membrane capacitance (C(m)) by whole-cell patch clamp in single JG cells as an index of exocytosis. Hypotonicity increased C(m) significantly and enhanced outward current. Indomethacin, PLA(2) inhibition, and an antagonist of prostaglandin transport impaired the C(m) and current responses to hypotonicity. Hypotonicity also increased exocytosis as determined by a decrease in single JG cell quinacrine fluorescence in an indomethacin-sensitive manner. In single JG cells from COX-2(-/ -) and AQP1(-/ -) mice, hypotonicity increased neither C(m) nor outward current, but 0.1-muM PGE(2) increased both in these cells. A reduction in osmolality enhanced cAMP accumulation in JG cells but not in renin-producing As4.1 cells; only the former had detectable AQP1 expression. Inhibition of protein kinase A blocked the hypotonicity-induced C(m) and current response in JG cells. Taken together, our results show that a 5 to 7% decrease in extracellular tonicity leads to AQP1-mediated water influx in JG cells, PLA(2)/COX-2-mediated prostaglandin-dependent formation of cAMP, and activation of PKA, which promotes exocytosis of renin.


Assuntos
Aquaporina 1/fisiologia , Ciclo-Oxigenase 2/fisiologia , Exocitose , Sistema Justaglomerular/metabolismo , Renina/metabolismo , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Soluções Hipotônicas , Sistema Justaglomerular/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Concentração Osmolar , Fosfolipases A2/fisiologia , Ratos , Ratos Sprague-Dawley
7.
J Hypertens ; 27(4): 829-37, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19516182

RESUMO

BACKGROUND: Preeclampsia is a serious pregnancy complication, accompanied by increased maternal and fetal morbidity. Different models have been used to study preeclampsia, but none of these display all the key features of the disease. METHOD: We investigated the effects on maternal blood pressure and fetal outcome exerted by the angiogenesis inhibitor Suramin (100 mg/kg i.p.) during early placentation. Blood pressure and heart rate were measured continuously with telemetry in Sprague-Dawley rats of four experimental groups: nonpregnant controls, Suramin-treated nonpregnant rats, pregnant controls and pregnant Suramin-treated rats. Blood samples were collected before pregnancy and at gestational day 20 for analysis of renin and sFlt-1. The fetal and placental morphology were evaluated after caesarian section on gestational day 20. RESULTS: The blood pressure of the pregnant Suramin-treated rats successively increased during pregnancy and differed by 17 mmHg at gestational day 20 compared with the pregnant control rats. In the pregnant Suramin-treated rats group, the renin levels increased (+122%) and the sFlt-1 levels decreased (-58%) during pregnancy. The pregnant Suramin-treated fetuses and placentae were smaller (2.8 g and 0.51 g) than the pregnant controls rats' fetuses and placentae (3.5 g and 0.56 g). Resorptions tended to be higher in the pregnant Suramin-treated rat litters compared with the pregnant control rat litters (P = 0.08). The area of the maternal blood vessels in the mesometrial triangle was smaller in the pregnant Suramin-treated rats group than in the pregnant control rats group. CONCLUSION: The inhibition of uterine angiogenesis increases maternal blood pressure and compromises fetal and placental development. Placental hypoxia and subsequent activation of the renin-angiotensin system may play an important role for the hypertension.


Assuntos
Inibidores da Angiogênese/toxicidade , Hipertensão/etiologia , Placenta/efeitos dos fármacos , Pré-Eclâmpsia/etiologia , Suramina/toxicidade , Útero/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Feminino , Frequência Cardíaca/efeitos dos fármacos , Masculino , Placenta/patologia , Placenta/fisiologia , Gravidez , Resultado da Gravidez , Ratos , Ratos Sprague-Dawley , Renina/sangue , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Útero/irrigação sanguínea , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/análise
8.
Kidney Int ; 75(8): 793-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19165174

RESUMO

Adenosine can induce vasodilatation and vasoconstriction of the renal afferent arteriole of the mouse. We determined here its direct effect on efferent arterioles of mouse kidneys. Using isolated-perfused cortical efferent arterioles, we measured changes in luminal diameter in response to adenosine. Extraluminal application of adenosine and cyclohexyladenosine had no effect on the luminal diameter. When the vessels were constricted by the thromboxane mimetic U46619, application of adenosine and 5'-N-ethylcarboxamido-adenosine dilated the efferent arterioles in a dose-dependent manner. We also found that the adenosine-induced vasodilatation was inhibited by the A(2)-specific receptor blocker 3,7-dimethyl-1-propargylxanthine. In the presence of this inhibitor, adenosine failed to alter the basal vessel diameter of quiescent efferent arterioles. Using primer-specific polymerase chain reaction we found that the adenosine A(1), A(2a), A(2b), and A(3) receptors were expressed in microdissected mouse efferent arterioles. We conclude that adenosine dilates the efferent arteriole using the A(2) receptor subtype at concentrations compatible with activation of the A(2b) receptor.


Assuntos
Córtex Renal/irrigação sanguínea , Receptores A2 de Adenosina/fisiologia , Vasodilatação , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Arteríolas/química , Relação Dose-Resposta a Droga , Técnicas In Vitro , Camundongos , Reação em Cadeia da Polimerase , Receptor A1 de Adenosina/análise , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/análise , Receptor A2A de Adenosina/genética , Receptor A2B de Adenosina/análise , Receptor A2B de Adenosina/genética , Receptor A3 de Adenosina/análise , Receptor A3 de Adenosina/genética , Receptores A2 de Adenosina/metabolismo , Vasodilatação/efeitos dos fármacos
9.
Physiology (Bethesda) ; 22: 310-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17928544

RESUMO

The aspartyl-protease renin is the key regulator of the renin-angiotensin-aldosterone system, which is critically involved in salt, volume, and blood pressure homeostasis of the body. Renin is mainly produced and released into circulation by the so-called juxtaglomerular epithelioid cells, located in the walls of renal afferent arterioles at the entrance of the glomerular capillary network. It has been known for a long time that renin synthesis and secretion are stimulated by the sympathetic nerves and the prostaglandins and are inhibited in negative feedback loops by angiotensin II, high blood pressure, salt, and volume overload. In contrast, the events controlling the function of renin-secreting cells at the organ and cellular level are markedly less clear and remain mysterious in certain aspects. The unravelling of these mysteries has led to new and interesting insights into the process of renin release.


Assuntos
Processamento de Proteína Pós-Traducional/fisiologia , Renina/biossíntese , Renina/metabolismo , Animais , AMP Cíclico/metabolismo , Humanos , Sistema Justaglomerular/citologia , Sistema Justaglomerular/metabolismo , Rim/citologia , Rim/metabolismo , Comunicação Parácrina/fisiologia , Vesículas Secretórias/metabolismo , Transdução de Sinais/fisiologia
11.
J Hypertens ; 24(10): 2001-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16957560

RESUMO

OBJECTIVE: Using an aortic constriction model in mice, we studied whether the increase in pressure or the activation of the renin-angiotensin system (RAS) and its main receptors is the main driving force for plaque progression. METHODS: Male ApoE mice underwent sham surgery or placement of a suprarenal silver clip around the aorta (AoC). Half the group was treated with the selective AT1 receptor antagonist losartan (30 mg/kg per day) for 4 weeks. RESULTS: Anesthetized mean arterial pressure (MAP) was increased in AoC mice compared to sham (106 +/- 3 versus 90 +/- 1 mmHg, P < 0.001). Losartan reduced MAP in sham mice (78 +/- 2 mmHg, P < 0.01) but not in AoC (AoC losartan 104 +/- 2 mmHg). Plasma renin concentration (PRC) was increased in AoC mice compared to sham [1.6 +/- 0.3 versus 0.8 +/- 0.2 milliGoldblatt units (mGU)/ml, P < 0.001]. Losartan treatment augmented this difference (18.7 +/- 3.7 versus 4.6 +/- 1.7 mGU/ml, P < 0.01). AT2 receptor mRNA expression was increased 5.8-fold by aortic constriction in thoracic aorta (P < 0.05) and the major site for expression of the AT2 receptor protein was within the plaques. The plaque area was increased in AoC mice compared to sham (0.61 +/- 0.09 versus 0.07 +/- 0.01%, P < 0.001); however, losartan did not alter plaque area. CONCLUSIONS: Our data do not support a role for the AT1 receptor in the progression of atherosclerosis in this model, since blockade with losartan did not alter plaque distribution. Furthermore, we found no support for the counteraction of atherogenesis by increased activity of the RAS acting on the AT2 receptor. Our data suggest that increased pressure is the main driving force for atherosclerosis in this model.


Assuntos
Estenose da Valva Aórtica/fisiopatologia , Aterosclerose/etiologia , Aterosclerose/fisiopatologia , Pressão Sanguínea/fisiologia , Receptor Tipo 1 de Angiotensina/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Estenose da Valva Aórtica/metabolismo , Aterosclerose/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Losartan/farmacologia , Masculino , Camundongos , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos
12.
Cardiovasc Res ; 71(3): 557-65, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16831408

RESUMO

OBJECTIVE: Extracellular retention of PDGF-B has been proposed to play an important role in PDGF-B signalling. We used the PDGF-B retention motif knockout mouse (RetKO) to study the effects of retention motif deletion on development of micro- and macrovascular structure and function. METHODS: Passive and active properties of conduit vessels were studied using myograph techniques and histological examination. Capillary structure and function was studied using measurements of capillary density in skeletal muscle and by assessing aerobic physical performance in a treadmill setup. Cardiac function was assessed using echocardiography. RESULTS: Myograph experiments revealed an increased diameter and stiffness of the aorta in RetKO. Histological examination showed increased media collagen content and a decreased number of aortic wall layers, however with a similar number of vascular smooth muscle cells. This outward eutrophic remodelling of the aorta was accompanied by endothelial dysfunction. RetKO showed decreased capillary density in skeletal muscle and signs of a defective delivery of capillary oxygen to skeletal muscle, as shown by a decreased physical performance. In RetKO mice, echocardiography revealed an adaptive eccentric cardiac hypertrophy. CONCLUSION: We conclude that retention of PDGF-B during development is essential for a normal conduit vessel function in the adult mouse. Furthermore, PDGF-B retention is also necessary for the development of an adequate capillary density, and thereby for a normal oxygen delivery to skeletal muscle. The lack of primary effects on cardiac function supports the redundant role of PDGF-B in cardiac development.


Assuntos
Aorta Torácica/fisiopatologia , Músculo Esquelético/irrigação sanguínea , Proteínas Proto-Oncogênicas c-sis/fisiologia , Animais , Aorta Torácica/patologia , Pressão Sanguínea , Capilares/patologia , Capilares/fisiopatologia , Cardiomegalia/diagnóstico , Cardiomegalia/genética , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora , Miografia , Consumo de Oxigênio , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/genética , Renina/sangue , Túnica Média/metabolismo
13.
Clin Sci (Lond) ; 111(1): 61-70, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16515534

RESUMO

The peptide AM (adrenomedullin) is stimulated by hypoxia through HIF-1 (hypoxia-inducible factor-1). The majority of human CC-RCCs (clear cell renal cell carcinomas) display mutations in the tumour suppressor protein von Hippel-Lindau, which leads to constitutively elevated HIF-1. We hypothesized that AM is increased in CC-RCC tumours and that AM is a plasma biomarker for CC-RCC. Tumours and non-malignant kidney tissue were obtained from patients that underwent unilateral nephrectomy. Blood samples were drawn at the day of surgery, 3-6 days after surgery and 4-5 weeks after surgery. AM mRNA and peptide expression in tissue and AM plasma concentration were determined. HIF-1alpha was localized in tissue by immunohistochemistry. AM mRNA was elevated in CC-RCC compared with adjacent renal cortex (6-fold, n=18; P<0.02). There was no difference in AM mRNA between cortex and non-CC-RCC tissue (n=7). AM peptide concentration was elevated in CC-RCC tissue compared with adjacent cortex (4-fold, n=6; P<0.02), whereas there was no difference between cortex and non-CC-RCC tissue (n=5). HIF-1alpha immunoreactivity was detected in the majority of cell nuclei in 76% of CC-RCC, consistent with constitutive stabilization. In non-CC-RCC, HIF-1alpha staining was focal. Before surgery there was no difference in plasma AM concentration between tumour types. Nephrectomy increased plasma AM significantly after 3-6 days and a similar pre-surgery level was observed after 4-5 weeks in both groups of tumour patients. We conclude that elevated tissue AM is a distinguishing feature of CC-RCC compared with other kidney tumours. Plasma AM is not suited as a tumour marker for this disease.


Assuntos
Adenocarcinoma de Células Claras/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Peptídeos/metabolismo , Adenocarcinoma de Células Claras/diagnóstico , Adenocarcinoma de Células Claras/cirurgia , Adrenomedulina , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/diagnóstico , Carcinoma de Células Renais/cirurgia , Diagnóstico Diferencial , Feminino , Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Córtex Renal/metabolismo , Neoplasias Renais/diagnóstico , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Nefrectomia , Peptídeos/sangue , Peptídeos/genética , Período Pós-Operatório , RNA Mensageiro/genética , RNA Neoplásico/genética , Radioimunoensaio/métodos , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
14.
Am J Physiol Regul Integr Comp Physiol ; 290(5): R1324-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16357099

RESUMO

The present study was performed to investigate the role of adenosine A1 receptors in regulating blood pressure in conscious mice. Adenosine A1-receptor knockout (A1R-/-) mice and their wild-type (A1R+/+) littermates were placed on standardized normal-salt (NS), high-salt (HS), or salt-deficient (SD) diets for a minimum of 10 days before telemetric blood pressure and urinary excretion measurements in metabolic cages. On the NS diet, daytime and nighttime mean arterial blood pressure (MAP) was 7-10 mmHg higher in A1R-/- than in A1R+/+ mice. HS diet did not affect the MAP in A1R-/- mice, but the daytime and nighttime MAP of the A1R+/+ mice increased by approximately 10 mmHg, to the same level as that in the A1R-/-. On the SD diet, day- and nighttime MAP decreased by approximately 6 mmHg in both A1R-/- and A1R+/+ mice, although the MAP remained higher in A1R-/- than in A1R+/+ mice. Although plasma renin levels decreased with increased salt intake in both genotypes, the A1R-/- mice had an approximately twofold higher plasma renin concentration on all diets compared with A1R+/+ mice. Sodium excretion was elevated in the A1R-/- compared with the A1R+/+ mice on the NS diet. There was no difference in sodium excretion between the two genotypes on the HS diet. Even on the SD diet, A1R-/- mice had an increased sodium excretion compared with A1R+/+ mice. An abolished tubuloglomerular feedback response and reduced tubular reabsorption can account for the elevated salt excretion found in A1R-/- animals. The elevated plasma renin concentrations found in the A1R-/- mice could also result in increased blood pressure. Our results confirm that adenosine, acting through the adenosine A1 receptor, plays an important role in regulating blood pressure, renin release, and sodium excretion.


Assuntos
Pressão Sanguínea/fisiologia , Receptor A1 de Adenosina/fisiologia , Renina/metabolismo , Animais , Dieta , Dieta Hipossódica , Diurese/efeitos dos fármacos , Feminino , Camundongos , Camundongos Knockout , Receptor A1 de Adenosina/genética , Renina/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cloreto de Sódio na Dieta/farmacologia , Telemetria , Urodinâmica , Equilíbrio Hidroeletrolítico/fisiologia
15.
Am J Physiol Renal Physiol ; 289(5): F989-97, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15985651

RESUMO

PGE(2) and PGI(2) stimulate renin secretion and cAMP accumulation in juxtaglomerular granular (JG) cells. We addressed, at the single-cell level, the receptor subtypes and intracellular transduction mechanisms involved. Patch clamp was used to determine cell capacitance (C(m)), current, and membrane voltage in response to PGE(2), EP2 and EP4 receptor agonists, and an IP receptor agonist. PGE(2) (0.1 micromol/l) increased C(m) significantly, and the increase was abolished by intracellular application of the protein kinase A antagonist Rp-8-CPT-cAMPS. EP2-selective ligands butaprost (1 micromol/l), AE1-259-01 (1 nmol/l), EP4-selective agonist AE1-329 (1 nmol/l), and IP agonist iloprost (1 micromol/l) significantly increased C(m) mediated by PKA. The EP4 antagonist AE3-208 (10 nmol/l) blocked the effect of EP4 agonist but did not alter the response to PGE(2). Application of both EP4 antagonist and EP2-antagonist AH-6809 abolished the effects of PGE(2) on C(m) and current. EP2 and EP4 ligands stimulated cAMP formation in JG cells. PGE(2) rapidly stimulated renin secretion from superfused JG cells and diminished the membrane-adjacent granule pool as determined by confocal microscopy. The membrane potential hyperpolarized significantly after PGE(2), butaprost, AE1-329 and AE1-259 and outward current was augmented in a PKA-dependent fashion. PGE(2)-stimulated outward current, but not C(m) change, was abolished by the BK(Ca) channel inhibitor iberiotoxin (300 nmol/l). EP2 and EP4 mRNA was detected in sampled JG cells, and the preglomerular and glomerular vasculature was immunopositive for EP4. Thus IP, EP2, and EP4 receptors are associated with JG cells, and their activation leads to rapid PKA-mediated exocytotic fusion and release of renin granules.


Assuntos
AMP Cíclico/fisiologia , Sistema Justaglomerular/fisiologia , Receptores de Prostaglandina E/fisiologia , Renina/farmacocinética , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Exocitose , Sistema Justaglomerular/citologia , Masculino , Potenciais da Membrana , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP4
17.
J Am Soc Nephrol ; 15(5): 1189-98, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15100359

RESUMO

The study was performed to elucidate the distribution and cellular localization of cyclooxygenase (COX)-2 in human kidney and to address localization of downstream targets for COX-derived prostanoids. Cortex and outer and inner medulla tissue were obtained from control kidneys (cancer specimens), kidneys with arterial stenosis, and kidneys of patients who received angiotensin II inhibition or acetylsalicylic acid. Ribonuclease protection assay and Western blot test revealed that COX-1 and -2 mRNA and protein were expressed in all regions of human kidney (mRNA ratio, cortex:outer medulla:inner medulla COX-1 1:3:20 and COX-2 1:1:3). In adult kidney, immunohistochemical labeling for COX-2 was associated with smooth muscle cells in pre- and postglomerular vessels and with endothelium, particularly in vasa recta and medullary capillaries. Western blot test confirmed COX-2 expression in renal artery. COX-2 had a similar localization in fetal kidney and was additionally observed in Henle's loop and macula densa. Human tissue arrays displayed COX-2 labeling of vascular smooth muscle in multiple extrarenal tissues. Vascular COX-2 expression was significantly increased in kidneys with arterial stenosis. COX-1 was colocalized with microsomal prostaglandin E(2) synthase (PGES) in collecting ducts, and PGES was also detected in macula densa cells. Vascular COX-2 was colocalized with prostaglandin E(2) EP4 receptors but not with EP2 receptors. Thus, renovascular COX-2 expression was a constitutive feature encountered in human kidneys at all ages, whereas COX-2 was seen in macula densa only in fetal kidney. Vascular COX-2 activity in human kidney and extrarenal tissues may support blood flow and affect vascular wall-blood interaction.


Assuntos
Endotélio Vascular/enzimologia , Isquemia/fisiopatologia , Isoenzimas/genética , Rim/irrigação sanguínea , Prostaglandina-Endoperóxido Sintases/genética , Receptores de Prostaglandina E/genética , Adulto , Angiotensina II/antagonistas & inibidores , Células Cultivadas , Ciclo-Oxigenase 2 , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Inibidores Enzimáticos/farmacologia , Feto , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Imuno-Histoquímica , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Rim/embriologia , Proteínas de Membrana , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Mensageiro/análise , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP4 , Obstrução da Artéria Renal/metabolismo , Obstrução da Artéria Renal/fisiopatologia , Artérias Umbilicais/citologia , Artérias Umbilicais/embriologia , Artérias Umbilicais/enzimologia
19.
Am J Physiol Regul Integr Comp Physiol ; 286(5): R942-51, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14715486

RESUMO

To investigate regional aspects of hypoxic regulation of adrenomedullin (AM) in kidneys, we mapped the distribution of AM in the rat kidney after hypoxia (normobaric hypoxic hypoxia, carbon monoxide, and CoCl(2) for 6 h), anemia (hematocrit lowered by bleeding) and after global transient ischemia for 1 h (unilateral renal artery occlusion and reperfusion for 6 and 24 h) and segmental infarct (6 and 24 h). AM expression and localization was determined in normal human kidneys and in kidneys with arterial stenosis. Hypoxia stimulated AM mRNA expression significantly in rat inner medulla (CO 13 times, 8% O(2) 6 times, and CoCl(2) 8 times), followed by the outer medulla and cortex. AM mRNA level was significantly elevated in response to anemia and occlusion-reperfusion. Immunoreactive AM was associated with the thin limbs of Henle's loop, distal convoluted tubule, collecting ducts, papilla surface epithelium, and urothelium. AM labeling was prominent in the inner medulla after CO and in the outer medulla after occlusion-reperfusion. The infarct border zone was strongly labeled for AM. In cultured inner medullary collecting duct cells, AM mRNA was significantly increased by hypoxia. AM mRNA was equally distributed in human kidney and AM was localized as in the rat kidney. In human kidneys with artery stenosis, AM mRNA was not significantly enhanced compared with controls, but AM immunoreactivity was observed in tubules, vessels, and glomerular cells. In summary, AM expression was increased in the rat kidney in response to hypoxic and ischemic hypoxia in keeping with oxygen gradients. AM was widely distributed in the human kidney with arterial stenosis. AM may play a significant role to counteract hypoxia in the kidney.


Assuntos
Hipóxia/fisiopatologia , Isquemia/fisiopatologia , Rim/fisiologia , Peptídeos/genética , Obstrução da Artéria Renal/fisiopatologia , Adrenomedulina , Anemia/metabolismo , Anemia/fisiopatologia , Animais , Células Cultivadas , Expressão Gênica , Humanos , Hipertensão Renal/metabolismo , Hipertensão Renal/fisiopatologia , Hipóxia/metabolismo , Imuno-Histoquímica , Isquemia/metabolismo , Rim/irrigação sanguínea , Rim/citologia , Masculino , Peptídeos/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Obstrução da Artéria Renal/metabolismo
20.
Am J Physiol Renal Physiol ; 285(2): F348-58, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12842861

RESUMO

Data suggest that mineralocorticoid selectivity is differentially regulated in epithelial target tissues. We investigated whether the level of dietary NaCl intake influenced the expression and tissue distribution of 11-beta-hydroxysteroid dehydrogenase type 2 (11betaHSD-2), aldosterone receptor (MR), and glucocorticoid receptor (GR) in rat colon, kidney, and cardiovascular tissue. Rats were fed a diet with 0.01 or 3% NaCl for 10 days. Messenger RNAs were analyzed with ribonuclease protection assay, 11betaHSD-2 protein by Western blot analysis, and localization of GR and 11betaHSD-2 by immunohistochemistry. NaCl restriction elevated plasma renin and aldosterone concentration, whereas corticosterone was unaltered. In distal colon, 11betaHSD-2 mRNA and protein were augmented significantly by low-NaCl intake and immunolabeling was widely distributed in crypt and surface epithelium. The MR mRNA level was decreased, whereas GR mRNA was unaltered in distal colon. MR, GR, and 11betaHSD-2 mRNAs were not changed in kidney cortex and medulla, left cardiac ventricle, and aorta. Immunofluorescence labeling showed that GR and 11betaHSD-2 localization was mutually exclusive in kidney. In colon epithelium, nuclear staining for GR subsided as perinuclear 11betaHSD-2 immunoreactivity increased with NaCl restriction. As a functional correlate of increased 11betaHSD-2 expression in colon, the GR-stimulated sodium-hydrogen exchanger NHE-3 was lowered by NaCl restriction. Inhibition of 11betaHSD-2 activity by carbenoxolone during NaCl restriction stimulated NHE-3 expression in colon. Dexamethasone stimulated NHE-3 both in colon and kidney. These data indicate that mineralocorticoid selectivity is physiologically regulated by NaCl intake at the level of 11betaHSD-2 expression and tissue distribution in the distal colon, but not in the kidney.


Assuntos
Colo/enzimologia , Hidroxiesteroide Desidrogenases/genética , Hidroxiesteroide Desidrogenases/metabolismo , Córtex Renal/enzimologia , Cloreto de Sódio na Dieta/farmacologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2 , Aldosterona/sangue , Animais , Sistema Cardiovascular/enzimologia , Corticosterona/sangue , Células Epiteliais/enzimologia , Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Renina/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA