Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Handb Exp Pharmacol ; (200): 391-412, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20859805

RESUMO

This chapter describes the effects of the natural methylxanthines caffeine and theophylline on kidney function. Theophylline in particular was used traditionally to increase urine out put until more potent diuretics became available in the middle of the last century. The mildly diuretic actions of both methylxanthines are mainly the result of inhibition of tubular fluid reabsorption along the renal proximal tubule. Based upon the use of specific adenosine receptor antagonists and the observation of a complete loss of diuresis in mice with targeted deletion of the A1AR gene, transport inhibition by methylxanthines is mediated mainly by antagonism of adenosine A1 receptors (A1AR) in the proximal tubule. Methylxanthines are weak renal vasodilators, and they act as competitive antagonists against adenosine-induced preglomerular vasoconstriction. Caffeine and theophylline stimulate the secretion of renin by inhibition of adenosine receptors and removal of the general inhibitory brake function of endogenous adenosine. Since enhanced intrarenal adenosine levels lead to reduced glomerular filtration rate in several pathological conditions theophylline has been tested for its therapeutic potential in the renal impairment following administration of nephrotoxic substances such as radiocontrast media, cisplatin, calcineurin inhibitors or following ischemia-reperfusion injury. In experimental animals functional improvements have been observed in all of these conditions, but available clinical data in humans are insufficient to affirm a definite therapeutic efficacy of methylxanthines in the prevention of nephrotoxic or postischemic renal injury.


Assuntos
Cafeína/farmacologia , Rim/efeitos dos fármacos , Teofilina/farmacologia , Animais , Meios de Contraste/toxicidade , Diurese/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Humanos , Rim/fisiologia , Natriurese/efeitos dos fármacos , Renina/metabolismo
2.
Pflugers Arch ; 460(1): 187-96, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20349193

RESUMO

AMP-activated protein kinase (AMPK) is expressed in the apical membrane of cortical thick ascending limb, distal, and collecting tubules as well as macula densa cells of the kidneys. AMPK is an active modulator of epithelial Na(+) channels, Na(+)-2Cl(-)-K(+) cotransporter, and the ATP-dependent potassium channel. The present experiments explored whether AMPK participates in the regulation of tubuloglomerular feedback (TGF) and renal tubular sodium handling. To this end, renal clearance and micropuncture experiments were performed in anesthetized rats. Under normal NaCl diet, neither TGF response nor renal fluid and sodium excretion were altered by pharmacological activation of AMPK in vivo. However, under high NaCl diet, the TGF response was significantly enhanced after intravenous or intratubular application of the AMPK activator AICAR. Moreover, AICAR application significantly increased fractional delivery of fluid and sodium to the end of the proximal tubule. High dietary NaCl intake increased the renal transcript levels encoding the AMPK-alpha1 subunit, while it decreased the expression of AMPK-beta1 and AMPK-gamma2 subunits. Immunoblots revealed that high dietary NaCl intake reduced renal expression of activated AMPK by about three times compared to normal NaCl diet whereas additional AICAR application increased AMPK activity. Our results suggest that AMPK regulates tubuloglomerular balance as well as tubular transport upon change of renal work load.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Glomérulos Renais/enzimologia , Túbulos Renais/enzimologia , Cloreto de Sódio na Dieta/metabolismo , Sódio/metabolismo , Equilíbrio Hidroeletrolítico , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/análogos & derivados , Animais , Ativação Enzimática , Ativadores de Enzimas/administração & dosagem , Retroalimentação Fisiológica , Hemodinâmica , Transporte de Íons , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Masculino , Fosforilação , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ribonucleotídeos/administração & dosagem , Sódio/urina , Cloreto de Sódio na Dieta/administração & dosagem , Urodinâmica , Equilíbrio Hidroeletrolítico/efeitos dos fármacos
3.
Cell Physiol Biochem ; 26(3): 449-56, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20798530

RESUMO

BACKGROUND/AIMS: Elevated plasma homocysteine (Hcy) levels have been identified as a pathogenic factor causing a variety of pathological changes in different cells and tissues. In vertebrates, Hcy is produced solely from S-adenosylhomocysteine (AdoHcy) through the catalysis of AdoHcy-hydrolase. The direction of AdoHcy-hydrolase activity is determined by its cytosolic substrate concentrations, thereby controlling intracellular AdoHcy levels. Most S-adenosylmethionine (AdoMet)-dependent methyltransferases are regulated in vivo by the ratio of AdoMet/AdoHcy, which is termed "methylation potential" (MP). To test whether high rates of erythropoietin (EPO) expression is reduced by a low MP in vivo we choosed the model of increased EPO production following carbon monoxide (CO) exposure in rats in which high transcriptional activity is responsible for renal EPO production. RESULTS: To induce a sustained hyperhomocysteinemia in rats, we infused i.v. a low or high dose of Hcy resulting in Hcy plasma levels of 87.4+/-6.2 and 300.8+/-23.7 mumol/l, respectively. Renal tissue contents of AdoHcy, AdoMet, and adenosine (Ado) were measured after freeze clamp by means of HPLC. Within 4h of CO exposure EPO serum levels increased from 13.6+/-0.4 (control) to 2254.8+/-278.3 mIU/ml. Only high dose of Hcy reduces both, the MP from 40.8+/-2.0 to 8.2+/-1.0 in the kidney as well as EPO serum levels by 40% compared to control rats. CONCLUSION: Our data show that severe hyperhomocysteinemia (HHcy) affects the MP in the renal tissue and lowers EPO expression following CO induced intoxication. This result supports the concept that efficient EPO production requires an unimpaired MP.


Assuntos
Eritropoetina/sangue , Hiper-Homocisteinemia/metabolismo , Adenosil-Homocisteinase/metabolismo , Animais , Monóxido de Carbono/toxicidade , Homocisteína/sangue , Hiper-Homocisteinemia/induzido quimicamente , Masculino , Ratos , Ratos Sprague-Dawley
4.
Handb Exp Pharmacol ; (193): 443-70, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19639291

RESUMO

The autacoid, adenosine, is present in the normoxic kidney and generated in the cytosol as well as at extracellular sites. The rate of adenosine formation is enhanced when the rate of ATP hydrolysis prevails over the rate of ATP synthesis during increased tubular transport work or during oxygen deficiency. Extracellular adenosine acts on adenosine receptor subtypes (A(1), A(2A), A(2B), and A(3)) in the cell membranes to affect vascular and tubular functions. Adenosine lowers glomerular filtration rate by constricting afferent arterioles, especially in superficial nephrons, and thus lowers the salt load and transport work of the kidney consistent with the concept of metabolic control of organ function. In contrast, it leads to vasodilation in the deep cortex and the semihypoxic medulla, and exerts differential effects on NaCl transport along the tubular and collecting duct system. These vascular and tubular effects point to a prominent role of adenosine and its receptors in the intrarenal metabolic regulation of kidney function, and, together with its role in inflammatory processes, form the basis for potential therapeutic approaches in radiocontrast media-induced acute renal failure, ischemia reperfusion injury, and in patients with cardiorenal failure.


Assuntos
Rim/fisiologia , Receptores Purinérgicos P1/fisiologia , Injúria Renal Aguda/tratamento farmacológico , Adenosina/uso terapêutico , Antagonistas do Receptor A1 de Adenosina , Animais , Taxa de Filtração Glomerular , Humanos , Receptor A1 de Adenosina/fisiologia , Renina/metabolismo , Teofilina/uso terapêutico
5.
Circulation ; 115(12): 1581-90, 2007 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-17353435

RESUMO

BACKGROUND: Ecto-5'-nucleotidase (CD73)-dependent adenosine generation has been implicated in tissue protection during acute injury. Once generated, adenosine can activate cell-surface adenosine receptors (A1 AR, A2A AR, A2B AR, A3 AR). In the present study, we define the contribution of adenosine to cardioprotection by ischemic preconditioning. METHODS AND RESULTS: On the basis of observations of CD73 induction by ischemic preconditioning, we found that inhibition or targeted gene deletion of cd73 abolished infarct size-limiting effects. Moreover, 5'-nucleotidase treatment reconstituted cd73-/- mice and attenuated infarct sizes in wild-type mice. Transcriptional profiling of adenosine receptors suggested a contribution of A2B AR because it was selectively induced by ischemic preconditioning. Specifically, in situ ischemic preconditioning conferred cardioprotection in A1 AR-/-, A2A AR-/-, or A3 AR-/- mice but not in A2B AR-/- mice or in wild-type mice after inhibition of the A2B AR. Moreover, A2B AR agonist treatment significantly reduced infarct sizes after ischemia. CONCLUSIONS: Taken together, pharmacological and genetic evidence demonstrate the importance of CD73-dependent adenosine generation and signaling through A2B AR for cardioprotection by ischemic preconditioning and suggests 5'-nucleotidase or A2B AR agonists as therapy for myocardial ischemia.


Assuntos
5'-Nucleotidase/fisiologia , Adenosina/fisiologia , Precondicionamento Isquêmico Miocárdico , Receptor A2B de Adenosina/fisiologia , 5'-Nucleotidase/deficiência , 5'-Nucleotidase/genética , Aminopiridinas/farmacologia , Aminopiridinas/uso terapêutico , Animais , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Hipóxia Celular/genética , Avaliação Pré-Clínica de Medicamentos , Líquido Extracelular/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/patologia , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/metabolismo , Neutrófilos , Receptor A2B de Adenosina/biossíntese , Receptor A2B de Adenosina/deficiência , Receptor A2B de Adenosina/efeitos dos fármacos , Receptor A2B de Adenosina/genética , Regulação para Cima , Xantinas/farmacologia , Xantinas/toxicidade
6.
PLoS Med ; 5(6): e137, 2008 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-18578565

RESUMO

BACKGROUND: Acute renal failure from ischemia significantly contributes to morbidity and mortality in clinical settings, and strategies to improve renal resistance to ischemia are urgently needed. Here, we identified a novel pathway of renal protection from ischemia using ischemic preconditioning (IP). METHODS AND FINDINGS: For this purpose, we utilized a recently developed model of renal ischemia and IP via a hanging weight system that allows repeated and atraumatic occlusion of the renal artery in mice, followed by measurements of specific parameters or renal functions. Studies in gene-targeted mice for each individual adenosine receptor (AR) confirmed renal protection by IP in A1(-/-), A2A(-/-), or A3AR(-/-) mice. In contrast, protection from ischemia was abolished in A2BAR(-/-) mice. This protection was associated with corresponding changes in tissue inflammation and nitric oxide production. In accordance, the A2BAR-antagonist PSB1115 blocked renal protection by IP, while treatment with the selective A2BAR-agonist BAY 60-6583 dramatically improved renal function and histology following ischemia alone. Using an A2BAR-reporter model, we found exclusive expression of A2BARs within the reno-vasculature. Studies using A2BAR bone-marrow chimera conferred kidney protection selectively to renal A2BARs. CONCLUSIONS: These results identify the A2BAR as a novel therapeutic target for providing potent protection from renal ischemia.


Assuntos
Citoproteção/genética , Isquemia/genética , Rim/irrigação sanguínea , Receptor A2B de Adenosina/fisiologia , Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina , Aminopiridinas/farmacologia , Animais , Vasos Sanguíneos/metabolismo , Citoproteção/efeitos dos fármacos , Líquido Extracelular/metabolismo , Feminino , Inflamação/genética , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Receptor A2B de Adenosina/genética , Receptor A2B de Adenosina/metabolismo , Transdução de Sinais/genética , Xantinas/farmacologia
7.
Cell Physiol Biochem ; 22(1-4): 223-36, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18769049

RESUMO

BACKGROUND/AIMS: S-Adenosylhomocysteine hydrolase (AdoHcyase) catalyzes the reversible hydrolysis of S-adenosylhomocysteine (AdoHcy), which is a potent product inhibitor of S-adenosylmethionine (AdoMet)-dependent methyltransferases. While previous studies have shown that AdoHcyase inhibition or deficiency lead to a decreased AdoMet/AdoHcy ratio resulting in impaired transmethylation, the effect of enhanced AdoHcyase activity on AdoMet/AdoHcy metabolism and methylation reactions has not been studied in detail. METHODS: To investigate the effect of enhanced AdoHcyase activity, we generated HEK-293 cell lines stably overexpressing AdoHcyase. RESULTS: Initial studies revealed that 2-10-fold AdoHcyase overexpression resulted in decreased intracellular AdoHcy and elevated adenosine levels, whereas 16-fold AdoHcyase overexpression increased adenosine and AdoHcy levels, lowered energy charge, and altered cell morphology. Furthermore, we found a correlation between AdoHcyase activity and cell viability. Caspase-activity assays and DNA fragmentation analysis revealed that the cell death in AdoHcyase overexpressing cells was due to apoptosis. Global DNA methylation was not altered in the different AdoHcyase overexpressing cell lines. CONCLUSION: Taken together, these data show that 2-5-fold enhanced AdoHcyase activity is well tolerated by the cell, while greatly enhanced AdoHcyase activity results in adenosine-induced apoptosis. The fact that enhanced AdoHcyase activity does not increase transmethylation activity suggests that AdoHcyase activity under physiological conditions is not rate limiting for efficient transmethylation.


Assuntos
Adenosina/metabolismo , Adenosil-Homocisteinase/metabolismo , Metilação de DNA , Espaço Intracelular/metabolismo , Adenosina Desaminase/metabolismo , Adenosil-Homocisteinase/genética , Caspases/metabolismo , Morte Celular , Linhagem Celular , Forma Celular , Sobrevivência Celular , Células Clonais , DNA/metabolismo , Fragmentação do DNA , Eletroforese em Gel de Ágar , Metabolismo Energético , Regulação Enzimológica da Expressão Gênica , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Frações Subcelulares , Transfecção
8.
FASEB J ; 21(11): 2863-73, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17442731

RESUMO

Previous studies showed increased extracellular nucleotides during renal ischemia-reperfusion. While nucleotides represent the main source for extracellular adenosine and adenosine signaling contributes to renal protection from ischemia, we hypothesized a role for ecto-nucleoside-triphosphate-diphosphohydrolases (E-NTPDases) in renal protection. We used a model of murine ischemia-reperfusion and in situ ischemic preconditioning (IP) via a hanging weight system for atraumatic renal artery occlusion. Initial studies with a nonspecific inhibitor of E-NTPDases (POM-1) revealed inhibition of renal protection by IP. We next pursued transcriptional responses of E-NTPDases (E-NTPDase1-3, and 8) to renal IP, and found a robust and selective induction of E-NTPDase1/CD39 transcript and protein. Moreover, based on clearance studies, plasma electrolytes, and renal tubular histology, IP protection was abolished in gene-targeted mice for cd39 whereas increased renal adenosine content with IP was attenuated. Furthermore, administration of apyrase reconstituted renal protection by IP in cd39-/- mice. Finally, apyrase treatment of wild-type mice resulted in increased renal adenosine concentrations and a similar degree of renal protection from ischemia as IP treatment. Taken together, these data identify CD39-dependent nucleotide phosphohydrolysis in renal protection. Moreover, the present studies suggest apyrase treatment as a novel pharmacological approach to renal diseases precipitated by limited oxygen availability.


Assuntos
Injúria Renal Aguda/prevenção & controle , Antígenos CD/fisiologia , Apirase/fisiologia , Precondicionamento Isquêmico , Rim/enzimologia , Traumatismo por Reperfusão/prevenção & controle , Injúria Renal Aguda/etiologia , Adenosina/metabolismo , Animais , Apirase/metabolismo , Feminino , Rim/patologia , Testes de Função Renal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Artéria Renal/enzimologia , Artéria Renal/patologia , Circulação Renal
9.
Kidney Blood Press Res ; 31(4): 280-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18753797

RESUMO

BACKGROUND/AIMS: The serum- and glucocorticoid-inducible kinase SGK1 was originally cloned as a glucocorticoid-regulated gene and later as a transcriptional target for mineralocorticoids. SGK1 regulates channels and transporters including the renal Na(+) channel ENaC. It contributes to mineralocorticoid regulation of renal Na(+) excretion and salt appetite. The present study explored the contribution of SGK1 to effects of glucocorticoids on mineral and electrolyte metabolism. METHODS: SGK1-knockout mice (sgk1(-/-)) and their wild-type littermates (sgk1(+/+)) were analyzed in metabolic cages with or without treatment for 14 days with dexamethasone (3 mg/kg b.w., i.p.). Blood pressure was determined by the tail-cuff method. RESULTS: Prior to treatment fluid intake, urinary flow rate, urinary Na(+), K(+), phosphate and Cl(-) excretion, plasma electrolyte and glucose concentrations as well as blood pressure were similar in sgk1(-/-) and sgk1(+/+) mice. Dexamethasone did not significantly alter renal Na(+), K(+), Cl(-) and Ca(2+) excretion but decreased plasma Ca(2+) and phosphate concentration in sgk1(+/+) mice. The effect on Ca(2+) was significantly augmented and the effect on phosphate significantly blunted in sgk1(-/-) mice. Dexamethasone significantly increased fasting blood glucose concentrations in both genotypes. Dexamethasone increased blood pressure in sgk1(+/+) mice, an effect significantly blunted in sgk1(-/-) mice. CONCLUSIONS: The present observations disclose SGK1-sensitive glucocorticoid effects on calcium-phosphate metabolism and blood pressure.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Eletrólitos/metabolismo , Glucocorticoides/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Glicemia/efeitos dos fármacos , Cálcio/metabolismo , Dexametasona/farmacologia , Rim/metabolismo , Camundongos , Camundongos Knockout , Fosfatos/metabolismo
10.
Diabetes ; 55(7): 2059-66, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16804076

RESUMO

Excess salt intake decreases peripheral glucose uptake, thus impairing glucose tolerance. Stimulation of cellular glucose uptake involves phosphatidylinositide-3-kinase (PI-3K)-dependent activation of protein kinase B/Akt. A further kinase downstream of PI-3K is serum- and glucocorticoid-inducible kinase (SGK)1, which is upregulated by mineralocorticoids and, thus, downregulated by salt intake. To explore the role of SGK1 in salt-dependent glucose uptake, SGK1 knockout mice (sgk1(-/-)) and their wild-type littermates (sgk1(+/+)) were allowed free access to either tap water (control) or 1% saline (high salt). According to Western blotting, high salt decreased and deoxycorticosterone acetate (DOCA; 35 mg/kg body wt) increased SGK1 protein abundance in skeletal muscle and fat tissue of sgk1(+/+) mice. Intraperitoneal injection of glucose (3 g/kg body wt) into sgk1(+/+) mice transiently increased plasma glucose concentration approaching significantly higher values ([glucose]p,max) in high salt (281 +/- 39 mg/dl) than in control (164 +/- 23 mg/dl) animals. DOCA did not significantly modify [glucose]p,max in control sgk1(+/+) mice but significantly decreased [glucose]p,max in high-salt sgk1(+/+) mice, an effect reversed by spironolactone (50 mg/kg body wt). [Glucose]p,max was in sgk1(-/-) mice insensitive to high salt and significantly higher than in control sgk1(+/+) mice. Uptake of 2-deoxy-d-[1,2-(3)H]glucose into skeletal muscle and fat tissue was significantly smaller in sgk1(-/-) mice than in sgk1(+/+) mice and decreased by high salt in sgk1(+/+) mice. Transfection of HEK-293 cells with active (S422D)SGK1, but not inactive (K127N)SGK, stimulated phloretin-sensitive glucose uptake. In conclusion, high salt decreases SGK1-dependent cellular glucose uptake. SGK1 thus participates in the link between salt intake and glucose tolerance.


Assuntos
Glicemia/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Desoxicorticosterona/farmacologia , Feminino , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Caracteres Sexuais , Espironolactona/farmacologia
11.
J Mol Med (Berl) ; 84(5): 396-404, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16604333

RESUMO

The mineralocorticoids aldosterone and deoxycorticosterone acetate (DOCA) stimulate renal tubular salt reabsorption, increase salt appetite, induce extracellular volume expansion, and elevate blood pressure. Cardiac effects of mineralocorticoids include stimulation of matrix protein deposition leading to cardiac fibrosis, which is at least partially due to the direct action of the hormones on cardiac cells. The signaling mechanisms mediating mineralocorticoid-induced cardiac fibrosis have so far remained elusive. Mineralocorticoids have been shown to upregulate the serum- and glucocorticoid-inducible kinase 1 (SGK1), which participates in the effects of mineralocorticoids on renal tubular Na+ reabsorption and salt appetite. To explore the involvement of SGK1 in the pathogenesis of mineralocorticoid-induced cardiac fibrosis, SGK1 knockout mice (sgk1-/-) and wild-type littermates (sgk1+/+) were implanted a 21-day-release 50-mg DOCA pellet and supplied with 1% NaCl in drinking water for 18 days. This DOCA/high-salt treatment increased blood pressure in both genotypes but led to significant cardiac fibrosis only in sgk1+/+ but not in sgk1-/- mice. According to real-time polymerase chain reaction and Western blotting, DOCA/high-salt treatment enhanced transcript levels and protein expression of cardiac connective tissue growth factor (CTGF) only in sgk1+/+ but not in sgk1-/- mice. Furthermore, DOCA (10 microM) upregulated CTGF expression and enhanced CTGF promoter activity in lung fibroblasts isolated from sgk1+/+ but not from sgk1-/- mice, an effect involving spironolactone-sensitive mineralocorticoid receptors and activation of nuclear factor-kappaB (NFkappaB). Our results suggest that SGK1 plays a decisive role in mineralocorticoid-induced CTGF expression and cardiac fibrosis.


Assuntos
Desoxicorticosterona/análogos & derivados , Cardiopatias/patologia , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Miocárdio/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Angiotensinas/efeitos dos fármacos , Angiotensinas/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo , Desoxicorticosterona/efeitos adversos , Fibrose/induzido quimicamente , Fibrose/metabolismo , Coração/efeitos dos fármacos , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Proteínas Imediatamente Precoces/efeitos dos fármacos , Proteínas Imediatamente Precoces/genética , Masculino , Camundongos , Camundongos Mutantes , Antagonistas de Receptores de Mineralocorticoides , Miocárdio/metabolismo , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Receptores de Mineralocorticoides/metabolismo , Sais/farmacologia , Espironolactona/farmacologia
13.
Trends Pharmacol Sci ; 25(6): 294-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15165742

RESUMO

S-Adenosylhomocysteine hydrolase (AdoHcyase) controls intracellular levels of S-adenosylhomocysteine (AdoHcy). AdoHcy is a potent product inhibitor of some S-adenosylmethionine-dependent methyltransferases. Pharmacological modulation of AdoHcyase to indirectly inhibit methyltransferases can be guided by the fact that adenosine binds with high affinity to AdoHcyase and inhibits enzyme activity. cAMP can compete with adenosine and can counteract the adenosine-induced inhibition of AdoHcyase. Thus, the ratio between adenosine and cAMP, which can vary under different physiological conditions, might result in changes in, for example, DNA promoter methylation and therefore transcription.


Assuntos
Adenosina/farmacologia , Adenosil-Homocisteinase/efeitos dos fármacos , Animais , AMP Cíclico/fisiologia , Humanos , Modelos Moleculares , S-Adenosil-Homocisteína/metabolismo
14.
Diabetes ; 53 Suppl 3: S128-34, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15561900

RESUMO

The sulfonylurea receptor (SUR) is the important regulatory subunit of ATP-sensitive K+ channels. It is an ATP-binding cassette protein comprising 17 transmembrane helices. SUR is endowed with binding sites for channel blockers like the antidiabetic sulfonylurea glibenclamide and for the chemically very heterogeneous channel openers. SUR1, the typical pancreatic SUR isoform, shows much higher affinity for glibenclamide but considerably lower affinity for most openers than SUR2. In radioligand binding assays, we investigated the role of two amino acids, T1285 and M1289, located in transmembrane helix (TM)-17, in opener binding to SUR1. These amino acids were exchanged for the corresponding amino acids of SUR2. In competition experiments using [3H]glibenclamide as radioligand, SUR1(T1285L, M1289T) showed much higher affinity toward the cyanoguanidine openers pinacidil and P1075 than SUR1 wild type. The affinity for the thioformamide aprikalim was also markedly increased. In contrast, the affinity for the benzopyrans rilmakalim and levcromakalim was unaffected; however, the amount of displaced [3H]glibenclamide binding was nearly doubled. The binding properties of the opener diazoxide and the blocker glibenclamide were unchanged. In conclusion, mutation of two amino acids in TM17 of SUR1, especially of M1289, leads to class-specific effects on opener binding by increasing opener affinity or by changing allosteric coupling between opener and glibenclamide binding.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Transportadores de Cassetes de Ligação de ATP , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Cromanos/farmacocinética , Cromakalim/farmacocinética , Diazóxido/farmacologia , Glibureto/farmacocinética , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Mutagênese Sítio-Dirigida , Oócitos , Conformação Proteica , Estrutura Secundária de Proteína , Pirrolidinas/farmacocinética , Ratos , Receptores de Droga , Proteínas Recombinantes/metabolismo , Receptores de Sulfonilureias , Transfecção , Xenopus
15.
Biochem Pharmacol ; 64(8): 1201-6, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12234600

RESUMO

The enzyme S-adenosyl-homocysteine hydrolase (AdoHcyase) which catalyzes the reversible hydrolysis of AdoHcy to adenosine and homocysteine is an adenosine binding protein. In the present study we examined the characteristics of [(3)H]cAMP binding to purified AdoHcyase from bovine kidney in comparison with the high affinity adenosine binding site of AdoHcyase. AdoHcyase exhibits one [(3)H]cAMP binding site with an affinity of K(d)=23.1+/-1.1nM and a B(max) of 116.6+/-3.8pmol/mg protein. Binding of [(3)H]cAMP obeyed a monophasic reaction with a k(+1) value of 0.035min/M. The dissociation of AdoHcyase-[(3)H]cAMP complex exhibited a time- and temperature-dependent character. After a 240min incubation at 0 degrees only 5-10%, however, at 20 degrees 90% were displaceable. Adenosine and cAMP displace each other with similar affinities of EC(50) 57nM vs. EC(50) 65nM. 2'-Deoxyadenosine, N(6)-methyladenosine, and NECA displace 25nM [(3)H]cAMP and 10nM [(3)H]adenosine with EC(50) values of 94, 90 and 80nM, respectively. All other nucleosides studied, adenine, inosine, adenosine-2',3'-dialdehyde, 2-chloroadenosine, aristeromycin, and adenine nucleotides were only week competitors for [(3)H]cAMP and [(3)H]adenosine. These compounds displace [(3)H]cAMP and [(3)H]adenosine with equal potencies. Our data indicate that the binding site for nanomolar concentrations of cAMP and adenosine at the AdoHcyase appears to be identical. The physiological implications of a cAMP binding site at the AdoHcyase remain to be established.


Assuntos
AMP Cíclico/metabolismo , Hidrolases/metabolismo , Rim/enzimologia , Adenosil-Homocisteinase , Animais , Sítios de Ligação , Bovinos , Hidrolases/efeitos dos fármacos , Hipoxantina/metabolismo , Teofilina/farmacologia , Trítio , Xantinas/farmacologia
16.
Biochem Pharmacol ; 66(11): 2117-23, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14609736

RESUMO

S-Adenosylhomocysteine hydrolase (AdoHcy hydrolase) catalyzes the reversible hydrolysis of S-adenosylhomocysteine (AdoHcy) to adenosine (Ado) and homocysteine. On the basis of the kinetics of Ado binding to AdoHcy hydrolase we have shown that AdoHcy hydrolase binds Ado with different affinities [Kidney Blood Press. Res. 19 (1996) 100]. Since AdoHcy hydrolase in its totally reduced form binds Ado with high affinity we determined in the present study the Ado binding characteristics of purified AdoHcy hydrolase from bovine kidney (native form) and of reconstituted forms with defined NAD(+)/NADH ratios. AdoHcy hydrolase in its native form and at a ratio of 50% NAD(+) and 50% NADH exhibits two binding sites for Ado with a K(D1) of 9.2+/-0.6 nmol/L and a K(D2) of 1.4+/-0.1 micromol/L, respectively. Binding of Ado to AdoHcy hydrolase in its NADH form and in its NAD(+) form exhibits only one binding site with high affinity 48.3+/-2.7 nmol/L for the NADH form and with a low affinity of 4.9+/-0.3 micromol/L for the NAD(+) form. To identify these two Ado binding sites, AdoHcy hydrolase was covalently modified with [2-3H]-8-azido-Ado. After irradiation of the native AdoHcy hydrolase two different photolabeled peptides were isolated and identified as Asp(307)-Val(325) and Tyr(379)-Thr(410). When the reconstituted AdoHcy hydrolase in its NADH and in its NAD(+) form was irradiated with [2-3H]-8-azido-Ado only one peptide was identified as Asn(312)-Lys(318) from the NADH form and as Asp(391)-Ala(396) from the NAD(+) form. Based on the crystallographic data, the labeled peptide Asp(391)-Ala(396) (low affinity binding site), appears to belong to the catalytic domain of AdoHcy hydrolase, whereas the labeled peptide, identified as Asn(312)-Lys(318) (high affinity binding site), is located in the NAD domain. In conclusion, our data show that AdoHcy hydrolase has two different Ado binding sites which are dependent upon the enzyme-bound NAD(+)/NADH ratios.


Assuntos
Adenosina/metabolismo , Adenosil-Homocisteinase/metabolismo , NAD/metabolismo , Sequência de Aminoácidos/fisiologia , Animais , Sítios de Ligação/fisiologia , Bovinos , Dados de Sequência Molecular , Ligação Proteica/fisiologia
17.
Biochem Pharmacol ; 63(4): 809-15, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11992651

RESUMO

Most S-adenosylmethionine (AdoMet)-dependent methyltransferases are regulated in vivo by the AdoMet/S-adenosylhomocysteine (AdoHcy) ratio, also termed as "methylation potential." Since adenosine inhibits in vitro AdoHcy hydrolysis and since adenosine tissue levels increase during hypoxia, it can be predicted that AdoHcy levels may increase in the rat kidney in parallel of those of adenosine. Therefore, the present investigation was performed to assess changes of renal AdoHcy and AdoMet tissue contents during ischemia and after administration of adenosine and homocysteine or both in the ischemic rat kidney. In anesthetized rats ischemia of the kidney was induced by renal artery occlusion for various time intervals. Adenosine and homocysteine were infused into the renal artery of the ischemic kidney. To induce a hyperhomocysteinemia homocysteine was continuously infused. The kidneys were removed and immediately snap-frozen. Tissue contents of AdoHcy, AdoMet, adenosine and adenine nucleotides were analyzed by means of HPLC. Under normoxic condition the tissue contents of AdoHcy, AdoMet and adenosine were 0.7+/-0.05, 44.1+/-1.0 and 3.8+/-0.1nmol/g wet weight, respectively. Renal ischemia for 30min resulted in an increase of AdoHcy levels from 0.7+/-0.05 to 9.1+/-0.6nmol/g wet weight and in a dramatic decrease of the AdoMet/AdoHcy ratio and energy charge from 65.1+/-5.6 to 2.8+/-0.2 and from 0.87+/-0.01 to 0.25+/-0.01, respectively. Application of exogenous adenosine into the ischemic kidney did not result in further AdoHcy accumulation. However, when homocysteine was infused into the ischemic kidney, AdoHcy increased five-fold above control levels, during 5min ischemia. Systemic infusion of homocysteine leads to a reduction of the methylation potential also in the normoxic kidney. We conclude that (i) the methylation potential in the kidney is markedly reduced during ischemia, mainly due to accumulation of AdoHcy; (ii) elevation of AdoHcy tissue content during ischemia is the result of the inhibition of AdoHcy hydrolysis; (iii) homocysteine is rate limiting for AdoHcy synthesis in the ischemic kidney; (iv) under normoxic conditions hyperhomocysteinemia can affect the methylation potential in the renal tissue.


Assuntos
Homocisteína/farmacologia , Isquemia/metabolismo , Nefropatias/metabolismo , S-Adenosil-Homocisteína/metabolismo , Adenosina/metabolismo , Animais , Modelos Animais de Doenças , Hiper-Homocisteinemia/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , S-Adenosilmetionina/metabolismo
18.
Hypertens Res ; 26(10): 855-61, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14621190

RESUMO

Defective dopamine receptors may be involved in the development of hypertension. Recently, it has been shown that gene expression and function of the renal dopamine D3 receptor is impaired in salt-sensitive Dahl rats, a model of salt-dependent hypertension. Here, the functional response to D3 receptor activation was investigated in spontaneously hypertensive rats (SHR) and their normotensive Wistar-Kyoto rats (WKY). In addition, expression of the D3 receptor gene was studied in both rat strains. In clearance experiments, Ringer solution was infused at baseline in thiopental-anesthetized SHR and WKY (each n = 8), followed by an infusion of R(+)-7-hydroxy-dipropylaminotetralin (DPAT), a specific D3 receptor agonist. DPAT was infused in two consecutive doses of 0.01 and 0.1 microg/min per kg body weight. During the entire experiment mean arterial blood pressure was significantly higher (1.5-fold) in adult SHR when compared to age-matched WKY. In both groups DPAT infusion induced a similar dose-dependent increase in urinary flow rate and sodium excretion by a maximum of 2.3-fold and 3.5-fold, respectively. DPAT also increased the glomerular filtration rate in both SHR and WKY. Reverse transcription-polymerase chain reaction studies of whole kidney samples showed no significant differences between young prehypertensive and adult hypertensive SHR when compared to age-matched normotensive WKY. In summary, pharmacological dopamine D3 receptor activation induces a uniform renal response in SHR and WKY. Together with the similar D3 receptor gene expression in both rat strains, which is independent of age or blood pressure levels, the results do not support the notion that the dopamine D3 receptor system is involved in the pathogenesis of hypertension in the SHR model.


Assuntos
Hipertensão Renal/fisiopatologia , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Animais , Pressão Sanguínea , Expressão Gênica , Taxa de Filtração Glomerular , Hipertensão Renal/metabolismo , Rim/fisiopatologia , Masculino , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores de Dopamina D3 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Urina
19.
Naunyn Schmiedebergs Arch Pharmacol ; 367(5): 427-33, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12709781

RESUMO

The dopamine D(2)-like receptor agonist quinpirole has been reported to lower blood pressure. This effect appears to be mediated via activation of presynaptic D(2)-like receptors inhibiting the stimulated neural norepinephrine release. The aim of the present study was to investigate the role of renal nerves and the renin-angiotensin system (RAS) in the blood pressure lowering effect of quinpirole. Therefore, clearance experiments using different doses of quinpirole (0.3 to 100 microg/kg/min) were performed in thiopental-anesthetized rats with intact kidneys (INN) or 5 to 7 days after bilateral renal denervation (DNX). The functional involvement of the RAS in the blood pressure lowering effect of quinpirole was determined in rats pretreated with a subpressor dose of angiotensin II (10 microg/kg/min) or in rats pretreated with the angiotensin II (AT(1)) receptor antagonist losartan, in a subdepressor dose (10 microg/kg/min). Quinpirole dose-dependently decreased mean arterial blood pressure (MAP) by up to 29%. This blood pressure lowering effect of quinpirole was observed at lower doses in DNX rats when compared with INN animals (ED(50): 0.98 microg/kg/min in DNX vs. 6.02 microg/kg/min in INN animals). Quinpirole in a dose of 3 microg/kg/min, which did not affect MAP in vehicle treated INN rats, significantly reduced MAP in rats with losartan pretreatment. In DNX rats pretreated with angiotensin II the MAP-response to the infusion of 3 microg/kg/min quinpirole was clearly attenuated in comparison with untreated DNX animals. Our data show that stimulation of dopamine D(2)-like receptors dose-dependently decreased blood pressure, which was potentiated by both interruption of the renal innervation and AT(1) receptor blockade, while exogenous ANG II restored the enhancement of the blood pressure response to quinpirole. We conclude that the increased vasodilatory effect of quinpirole after renal denervation might depend on a decreased activity of the RAS.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hipotensão/induzido quimicamente , Quimpirol/farmacocinética , Sistema Renina-Angiotensina/fisiologia , Anestesia , Angiotensina II/administração & dosagem , Angiotensina II/farmacocinética , Animais , Denervação , Relação Dose-Resposta a Droga , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Hipotensão/fisiopatologia , Rim/inervação , Rim/fisiologia , Rim/cirurgia , Testes de Função Renal , Losartan/administração & dosagem , Losartan/farmacocinética , Masculino , Quimpirol/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos
20.
J Clin Invest ; 122(2): 693-710, 2012 02.
Artigo em Inglês | MEDLINE | ID: mdl-22269324

RESUMO

A complex biologic network regulates kidney perfusion under physiologic conditions. This system is profoundly perturbed following renal ischemia, a leading cause of acute kidney injury (AKI) - a life-threatening condition that frequently complicates the care of hospitalized patients. Therapeutic approaches to prevent and treat AKI are extremely limited. Better understanding of the molecular pathways promoting postischemic reflow could provide new candidate targets for AKI therapeutics. Due to its role in adapting tissues to hypoxia, we hypothesized that extracellular adenosine has a regulatory function in the postischemic control of renal perfusion. Consistent with the notion that equilibrative nucleoside transporters (ENTs) terminate adenosine signaling, we observed that pharmacologic ENT inhibition in mice elevated renal adenosine levels and dampened AKI. Deletion of the ENTs resulted in selective protection in Ent1-/- mice. Comprehensive examination of adenosine receptor-knockout mice exposed to AKI demonstrated that renal protection by ENT inhibitors involves the A2B adenosine receptor. Indeed, crosstalk between renal Ent1 and Adora2b expressed on vascular endothelia effectively prevented a postischemic no-reflow phenomenon. These studies identify ENT1 and adenosine receptors as key to the process of reestablishing renal perfusion following ischemic AKI. If translatable from mice to humans, these data have important therapeutic implications.


Assuntos
Injúria Renal Aguda/metabolismo , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Isquemia/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Adenosina/metabolismo , Animais , Linhagem Celular , Quimerismo , Dipiridamol/uso terapêutico , Transportador Equilibrativo 1 de Nucleosídeo/antagonistas & inibidores , Transportador Equilibrativo 1 de Nucleosídeo/genética , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenômeno de não Refluxo , Proteínas de Transporte de Nucleosídeos/antagonistas & inibidores , Proteínas de Transporte de Nucleosídeos/metabolismo , Inibidores de Fosfodiesterase/uso terapêutico , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA