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










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 5: 17316, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26611568

RESUMO

The mitogen- and stress-activated kinase MSK1/2 plays a decisive role in apoptosis. In analogy to apoptosis of nucleated cells, suicidal erythrocyte death called eryptosis is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine (PS) externalization. Here, we explored whether MSK1/2 participates in the regulation of eryptosis. To this end, erythrocytes were isolated from mice lacking functional MSK1/2 (msk(-/-)) and corresponding wild-type mice (msk(+/+)). Blood count, hematocrit, hemoglobin concentration and mean erythrocyte volume were similar in both msk(-/-) and msk(+/+) mice, but reticulocyte count was significantly increased in msk(-/-) mice. Cell membrane PS exposure was similar in untreated msk(-/-) and msk(+/+) erythrocytes, but was enhanced by pathophysiological cell stressors ex vivo such as hyperosmotic shock or energy depletion to significantly higher levels in msk(-/-) erythrocytes than in msk(+/+) erythrocytes. Cell shrinkage following hyperosmotic shock and energy depletion, as well as hemolysis following decrease of extracellular osmolarity was more pronounced in msk(-/-) erythrocytes. The in vivo clearance of autologously-infused CFSE-labeled erythrocytes from circulating blood was faster in msk(-/-) mice. The spleens from msk(-/-) mice contained a significantly greater number of PS-exposing erythrocytes than spleens from msk(+/+) mice. The present observations point to accelerated eryptosis and subsequent clearance of erythrocytes leading to enhanced erythrocyte turnover in MSK1/2-deficient mice.


Assuntos
Apoptose/genética , Eritrócitos/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Animais , Índices de Eritrócitos , Eritrócitos/patologia , Feminino , Expressão Gênica , Hematócrito , Hemoglobinas , Hemólise , Humanos , Masculino , Camundongos , Camundongos Knockout , Fragilidade Osmótica , Pressão Osmótica , Fosfatidilserinas/metabolismo , Cultura Primária de Células , Contagem de Reticulócitos , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência
2.
Cell Physiol Biochem ; 32(3): 766-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24080829

RESUMO

BACKGROUND: The economic burden associated with major depressive disorder and anxiety disorders render both disorders the most common and debilitating psychiatric illnesses. To date, the exact cellular and molecular mechanisms underlying the pathophysiology, successful treatment and prevention of these highly associated disorders have not been identified. Akt2 is a key protein in the phosphatidylinositide-3 (PI3K) / glycogen synthase 3 kinase (GSK3) signaling pathway, which in turn is involved in brain-derived neurotrophic factor (BDNF) effects on fear memory, mood stabilisation and action of several antidepressant drugs. The present study thus explored the impact of Akt2 on behaviour of mice. METHODS: Behavioural studies (Open-Field, Light-Dark box, O-Maze, Forced Swimming Test, Emergence Test, Object Exploration Test, Morris Water Maze, Radial Maze) have been performed with Akt2 knockout mice (akt(-/-)) and corresponding wild type mice (akt(+/+)). RESULTS: Anxiety and depressive behavior was significantly higher in akt(-/-) than in akt(+/+) mice. The akt(-/-) mice were cognitively unimpaired but displayed increased anxiety in several behavioral tests (O-Maze test, Light-Dark box, Open Field test). Moreover, akt(-/-) mice spent more time floating in the Forced Swimming test, which is a classical feature of experimental depression. CONCLUSION: Akt2 might be a key factor in the pathophysiology of depression and anxiety.


Assuntos
Ansiedade/fisiopatologia , Depressão/fisiopatologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
Cell Physiol Biochem ; 32(4): 838-48, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24081014

RESUMO

BACKGROUND: The serum and glucocorticoid inducible kinase SGK1, which was originally cloned from mammary tumor cells, is highly expressed in some but not all tumors. SGK1 confers survival to several tumor cells. Along those lines, the number of colonic tumors following chemical carcinogenesis was decreased in SGK1 knockout mice. Recently, a highly selective SGK inhibitor (EMD638683) has been developed. The present study explored whether EMD638683 affects survival of colon carcinoma cells in vitro and impacts on development of colonic tumors in vivo. METHODS: Colon carcinoma (Caco-2) cells were exposed to EMD638683 with or without exposure to radiation (3 Gray) and cell volume was estimated from forward scatter, phosphatidylserine exposure from annexin V binding, mitochondrial potential from JC-9 fluorescence, caspase 3 activity from CaspGlow Fluorescein staining, DNA degradation from propidium iodide staining as well as late apoptosis from annexin-V FITC and propidium iodide double staining. In vivo tumor growth was determined in wild type mice subjected to chemical carcinogenesis (intraperitoneal injection of 20 mg/kg 1,2-dimethylhydrazine followed by three cycles of 30 g/L synthetic dextran sulfate sodium in drinking water for 7 days). RESULTS: EMD638683 treatment significantly augmented the radiation-induced decrease of forward scatter, increase of phosphatidylserine exposure, decrease of mitochondrial potential, increase of caspase 3 activity, increase of DNA fragmentation and increase of late apoptosis. The in vivo development of tumors following chemical carcinogenesis was significantly blunted by treatment with EMD638683. CONCLUSIONS: EMD638683 promotes radiation-induced suicidal death of colon tumor cells in vitro and decreases the number of colonic tumors following chemical carcinogenesis in vivo.


Assuntos
Benzamidas/farmacologia , Benzamidas/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Hidrazinas/farmacologia , Hidrazinas/uso terapêutico , Proteínas Imediatamente Precoces/antagonistas & inibidores , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Células CACO-2 , Caspase 3/metabolismo , Citometria de Fluxo , Humanos , Camundongos
4.
Nat Med ; 19(7): 934-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23770692

RESUMO

Major depression is a highly prevalent severe mood disorder that is treated with antidepressants. The molecular targets of antidepressants require definition. We investigated the role of the acid sphingomyelinase (Asm)-ceramide system as a target for antidepressants. Therapeutic concentrations of the antidepressants amitriptyline and fluoxetine reduced Asm activity and ceramide concentrations in the hippocampus, increased neuronal proliferation, maturation and survival and improved behavior in mouse models of stress-induced depression. Genetic Asm deficiency abrogated these effects. Mice overexpressing Asm, heterozygous for acid ceramidase, treated with blockers of ceramide metabolism or directly injected with C16 ceramide in the hippocampus had higher ceramide concentrations and lower rates of neuronal proliferation, maturation and survival compared with controls and showed depression-like behavior even in the absence of stress. The decrease of ceramide abundance achieved by antidepressant-mediated inhibition of Asm normalized these effects. Lowering ceramide abundance may thus be a central goal for the future development of antidepressants.


Assuntos
Antidepressivos/farmacologia , Ceramidas/fisiologia , Esfingomielina Fosfodiesterase/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Células Cultivadas , Ceramidas/metabolismo , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/metabolismo , Embrião de Mamíferos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo
5.
Kidney Blood Press Res ; 35(5): 365-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22473073

RESUMO

BACKGROUND/AIMS: Gum arabic (GA) is a Ca(2+)-, Mg(2+)- and K(+)-rich dietary fiber used for the treatment of patients with chronic kidney disease in Middle Eastern countries. In healthy mice, GA treatment increases creatinine clearance, renal ADH excretion, as well as intestinal and renal excretion of Mg(2+) and Ca(2+). GA decreases plasma Pi concentration, urinary Pi and Na(+) excretion. The present study explored the effects of GA on renal function in diabetic mice. METHODS: Metabolic cage experiments were performed on Akita mice (akita(+/-)), which spontaneously develop insulin deficiency and thus hyperglycemia. Plasma and urinary concentrations of Na(+), K(+) and Ca(2+) were measured by flame photometry (AFM 5051, Eppendorf, Germany), creatinine by the Jaffé method, phosphate photometrically, urea by an enzymatic method, glucose utilizing a glucometer and an enzymatic kit, aldosterone using an RIA, urinary albumin fluorometrically, and blood pressure by the tail-cuff method. RESULTS: GA (10% in drinking water) significantly increased urinary excretion of Ca(2+) and significantly decreased plasma phosphate and urea concentrations, urinary flow rate, urinary Na(+), phosphate and glucose excretion, blood pressure and proteinuria. CONCLUSIONS: GA treatment decreases blood pressure and proteinuria in diabetic mice and may thus prove beneficial in diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Goma Arábica/farmacologia , Rim/efeitos dos fármacos , Rim/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Cálcio/sangue , Cálcio/urina , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Goma Arábica/química , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Hiperglicemia/fisiopatologia , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/metabolismo , Hipertensão Renal/fisiopatologia , Masculino , Camundongos , Camundongos Mutantes , Potássio/sangue , Potássio/urina , Proteinúria/tratamento farmacológico , Proteinúria/metabolismo , Proteinúria/fisiopatologia , Sódio/sangue , Sódio/urina
6.
Cell Physiol Biochem ; 28(1): 137-46, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21865856

RESUMO

UNLABELLED: The serum- and glucocorticoid-inducible kinase 1 (SGK1) is transcriptionally upregulated by mineralocorticoids and activated by insulin. The kinase enhances renal tubular Na(+)-reabsorption and accounts for blood pressure increase following high salt diet in mice made hyperinsulinemic by dietary fructose or fat. The present study describes the in vitro and in vivo efficacy of a novel SGK1 inhibitor (EMD638683). EMD638683 was tested in vitro by determination of SGK1-dependent phosphorylation of NDRG1 (N-Myc downstream-regulated gene 1) in human cervical carcinoma HeLa-cells. In vivo EMD638683 (4460 ppm in chow, i.e. approx. 600 mg/kg/day) was administered to mice drinking tap water or isotonic saline containing 10% fructose. Blood pressure was determined by the tail cuff method, and urinary electrolyte (flame photometry) concentrations determined in metabolic cages. In vitro testing disclosed EMD638683 as a SGK1 inhibitor with an IC50 of 3 µM. Within 24 hours in vivo EMD638683 treatment significantly decreased blood pressure in fructose/saline-treated mice but not in control animals or in SGK1 knockout mice. EMD638683 failed to alter the blood pressure in SGK1 knockout mice. Following chronic (4 weeks) fructose/high salt treatment, additional EMD638683 treatment again decreased blood pressure. EMD638683 thus abrogates the salt sensitivity of blood pressure in hyperinsulinism without appreciably affecting blood pressure in the absence of hyperinsulinism. EMD638683 tended to increase fluid intake and urinary excretion of Na(+), significantly increased urinary flow rate and significantly decreased body weight. CONCLUSION: EMD638683 could serve as a template for drugs counteracting hypertension in individuals with type II diabetes and metabolic syndrome.


Assuntos
Anti-Hipertensivos/farmacologia , Benzamidas/farmacologia , Hidrazinas/farmacologia , Proteínas Imediatamente Precoces/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Anti-Hipertensivos/química , Anti-Hipertensivos/uso terapêutico , Benzamidas/química , Benzamidas/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Frutose/farmacologia , Células HeLa , Humanos , Hidrazinas/química , Hidrazinas/uso terapêutico , Hipertensão/tratamento farmacológico , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Knockout , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Sódio/urina , Cloreto de Sódio/farmacologia
7.
J Am Soc Nephrol ; 22(5): 873-80, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21493770

RESUMO

Insulin and IGF1-dependent signaling activates protein kinase B and serum and glucocorticoid inducible kinase (PKB/SGK), which together phosphorylate and inactivate glycogen synthase kinase GSK3. Because insulin and IGF1 increase renal tubular calcium and phosphorus reabsorption, we examined GSK3 regulation of phosphate transporter activity and determined whether PKB/SGK inactivates GSK3 to enhance renal phosphate and calcium transport. Overexpression of GSK3 and the phosphate transporter NaPi-IIa in Xenopus oocytes decreased electrogenic phosphate transport compared with NaPi-IIa-expressing oocytes. PKB/SGK serine phosphorylation sites in GSK3 were mutated to alanine to create gsk3(KI) mice resistant to PKB/SGK inactivation. Compared with wildtype animals, gsk3(KI) animals exhibited greater urinary phosphate and calcium clearances with higher excretion rates and lower plasma concentrations. Isolated brush border membranes from gsk3(KI) mice showed less sodium-dependent phosphate transport and Na-phosphate co-transporter expression. Parathyroid hormone, 1,25-OH vitamin D levels, and bone mineral density were decreased in gsk3(KI) mice, suggesting a global dysregulation of bone mineral metabolism. Taken together, PKB/SGK phosphorylation of GSK3 increases phosphate transporter activity and reduces renal calcium and phosphate loss.


Assuntos
Cálcio/urina , Quinase 3 da Glicogênio Sintase/fisiologia , Hipofosfatemia Familiar/etiologia , Proteínas Imediatamente Precoces/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Densidade Óssea , Calcitriol/sangue , Túbulos Renais/metabolismo , Camundongos , Hormônio Paratireóideo/sangue , Fosfatos/metabolismo , Fosforilação
8.
Pflugers Arch ; 461(5): 527-36, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21476133

RESUMO

Loss of function mutations of the tumor suppressor gene adenomatous polyposis coli (APC) underly the familial adenomatous polyposis. Mice carrying an inactivating mutation in the apc gene (apc (Min/+)) similarly develop intestinal polyposis. APC is effective at least in part by degrading ß-catenin and lack of APC leads to markedly enhanced cellular ß-catenin levels. ß-Catenin has most recently been shown to upregulate the Na+/K+ ATPase. The present study, thus, explored the possibility that APC could influence intestinal transport. The abundance and localization of ß-catenin were determined utilizing Western blotting and confocal microscopy, the activity of the electrogenic glucose carrier (SGLT1) was estimated from the glucose-induced current in jejunal segments utilizing Ussing chamber experiments and the Na+/H+ exchanger (NHE3) activity from Na+ -dependent re-alkalinization of cytosolic pH (ΔpH(i)) following an ammonium pulse employing BCECF fluorescence. As a result, ß-catenin abundance in intestinal tissue was significantly higher in apc (Min/+) mice than in wild-type mice (apc (+/+)). The ß-catenin protein was localized in the basolateral membrane. Both, the glucose-induced current and ΔpH(i) were significantly higher in apc (Min/+) mice than in apc (+/+) mice. In conclusion, intestinal electrogenic transport of glucose and intestinal Na+/H+ exchanger activity are both significantly enhanced in apc (Min/+) mice, pointing to a role of APC in the regulation of epithelial transport.


Assuntos
Genes APC/fisiologia , Mucosa Intestinal/metabolismo , beta Catenina/metabolismo , Animais , Transporte Biológico/genética , Regulação para Baixo , Glucose/metabolismo , Concentração de Íons de Hidrogênio , Camundongos , Transportador 1 de Glucose-Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo
9.
Am J Physiol Renal Physiol ; 299(5): F1171-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20719979

RESUMO

Klotho is a membrane protein participating in the inhibitory effect of FGF23 on the formation of 1,25-dihydroxyvitamin-D(3) [1,25(OH)(2)D(3)]. It participates in the regulation of renal tubular phosphate reabsorption and stimulates renal tubular Ca(2+) reabsorption. Klotho hypomorphic mice (klotho(hm)) suffer from severe growth deficit, rapid aging, and early death, events largely reversed by a vitamin D-deficient diet. The present study explored the role of Klotho deficiency in mineral and electrolyte metabolism. To this end, klotho(hm) mice and wild-type mice (klotho(+/+)) were subjected to a normal (D(+)) or vitamin D-deficient (D(-)) diet or to a vitamin D-deficient diet for 4 wk and then to a normal diet (D(-/+)). At the age of 8 wk, body weight was significantly lower in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice, klotho(hm)D(-) mice, and klotho(hm)D(-/+) mice. Plasma concentrations of 1,25(OH)(2)D(3,) adrenocorticotropic hormone (ACTH), antidiuretic hormone (ADH), and aldosterone were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. Plasma volume was significantly smaller in klotho(hm)D(-/+) mice, and plasma urea, Ca(2+), phosphate and Na(+), but not K(+) concentrations were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. The differences were partially abrogated by a vitamin D-deficient diet. Moreover, the hyperaldosteronism was partially reversed by Ca(2+)-deficient diet. Ussing chamber experiments revealed a marked increase in amiloride-sensitive current across the colonic epithelium, pointing to enhanced epithelial sodium channel (ENaC) activity. A salt-deficient diet tended to decrease and a salt-rich diet significantly increased the life span of klotho(hm)D(+) mice. In conclusion, the present observation disclose that the excessive formation of 1,25(OH)(2)D(3) in Klotho-deficient mice results in extracellular volume depletion, which significantly contributes to the shortening of life span.


Assuntos
Glucuronidase/genética , Glucuronidase/fisiologia , Hiperaldosteronismo/genética , Hormônio Adrenocorticotrópico/sangue , Aldosterona/sangue , Animais , Análise Química do Sangue , Pressão Sanguínea/fisiologia , Peso Corporal/fisiologia , Calcitriol/metabolismo , Cultura em Câmaras de Difusão , Eletrólitos/metabolismo , Fator de Crescimento de Fibroblastos 23 , Hiperaldosteronismo/metabolismo , Proteínas Klotho , Camundongos , Camundongos Knockout , Hormônio Paratireóideo/sangue , Volume Plasmático/fisiologia , Sobrevida , Vasopressinas/sangue
10.
Nutr Cancer ; 62(6): 802-10, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20661830

RESUMO

Gum Arabic (GA), a nutrient from dried exudate of Acacia senegal, is widely used as emulsifier and stabilizer. It stimulates sodium and water absorption in diarrhea. This study explored the effects of GA in colonic tissue. Mice were treated with GA (10% wt/vol) in drinking water and gene array was performed. As GA modified several tumor-relevant genes, chemical cancerogenesis (intraperitoneal injection of 20 mg/kg 1,2-dimethylhydrazine followed by 3 cycles of 3% dextrane sodium sulphate in drinking water) was induced with or without GA treatment. Within 4 days, GA treatment decreased the colonic transcript levels of the angiogenetic factors angiogenin 1, angiogenin 3, and angiogenin 4 by 78 +/- 18%, 88 +/- 15%, and 92 +/- 13%, respectively (n = 5 each), and of further genes including CD38 antigen, aquaporin4, interleukin18, vav-3-oncogene, gamma(+)-amino acid transporter, sulfatase1, ubiquitinD, and chemokine ligand5. According to Western blotting, GA treatment similarly decreased angiogenin protein expression, and according to immunohistochemistry, it decreased ss-catenin expression. Chemical cancerogenesis resulted in multiple colonic tumors within 12 wk. GA treatment (10% wt/wt) in drinking water significantly decreased the number of tumors by 70%. The observations disclose a powerful anticarcinogenic effect of GA. The nutrient could thus be used for the prophylaxis against colon carcinoma particularly in individuals at enhanced risk.


Assuntos
Anticarcinógenos/farmacologia , Neoplasias do Colo/prevenção & controle , Goma Arábica/farmacologia , Ribonuclease Pancreático/antagonistas & inibidores , 1,2-Dimetilidrazina , Animais , Regulação para Baixo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Ribonuclease Pancreático/análise , Ribonuclease Pancreático/genética
11.
Cell Physiol Biochem ; 25(6): 775-86, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20511724

RESUMO

BACKGROUND: Mounting evidence suggests that bipolar disorder symptoms could be favorably influenced by modification of glycogen synthase kinase-3 (GSK-3) activity. Specifically, the well known antimanic and mood stabilizing medications lithium, valproate, olanzapine and clozapine have been shown to inhibit GSK-3 activity. GSK-3 is phosphorylated and thus inhibited by protein kinase B (PKB/Akt) and serum and glucocorticoid inducible kinase (SGK) isoforms. The present study explored, whether PKB/SGK-dependent GSK-3 regulation influences the behavior of mice. METHODS: Gene-targeted knockin mice with mutated and thus PKB/SGK-resistant GSK-3alpha, beta (gsk-3(KI)) were compared to corresponding wild type mice (gsk-3(WT)). The mice were analyzed by open-field, light-dark (LD-) box, O-maze, emergence test, object exploration test and forced swimming test (FST). RESULTS: In open-field, LD-box and O-maze, gsk-3(KI) mice displayed a hyperactive and more curious phenotype when compared to wild type mice. Speed and total distance moved as well as rearings were significantly increased in gsk-3(KI) compared to gsk-3(WT) mice. In the O-maze, gsk-3(KI) mice tended to travel a larger distance in the open, unprotected area thangsk-3(WT) mice, and performed significantly more unprotected head dips suggesting decreased anxiety behavior. In the forced swimming test, the immobility time was significantly decreased in gsk-3(KI) mice indicating a phenotype less prone to depression. Moreover, gsk-3(KI)mice were less sensitive to the application of chronic mild stress and showed a decreased HPA axis activity. CONCLUSIONS: The present observations disclose a significant role of PKB/SGK-dependent regulation of GSK-3 in the control of activity, anxiety and proneness to depression. Accordingly, mice expressing SGK/PKB resistant GSK-3 may be a valuable model of hyperactivity and mania.


Assuntos
Comportamento Exploratório , Quinase 3 da Glicogênio Sintase/metabolismo , Hipercinese/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Depressão/genética , Depressão/metabolismo , Feminino , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Hipercinese/genética , Proteínas Imediatamente Precoces/genética , Masculino , Camundongos , Mutação , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética
12.
Am J Physiol Renal Physiol ; 298(5): F1113-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164156

RESUMO

Akt/PKB is known to regulate the facilitative glucose carrier GLUT4. Nothing is known, however, of the role of Akt/PKB in the regulation of renal epithelial transport. To explore whether Akt2/PKBß influences the Na(+)-coupled glucose cotransporter SGLT1, human SGLT1 was expressed in Xenopus laevis oocytes with or without Akt/PKB, and electrogenic glucose transport was determined by dual-electrode voltage clamp. The coexpression of Akt/PKB in SGLT1-expressing oocytes was followed by an increase in glucose-induced currents. To study the functional significance of Akt/PKB-sensitive renal glucose transport, further experiments were performed in gene-targeted mice lacking functional Akt2/PKBß (akt2(-/-)) and in their wild-type littermates (akt2(+/+)). Plasma glucose concentration was significantly higher in akt2(-/-) mice than in akt2(+/+) mice but was virtually identical to the plasma glucose concentration in fructose-treated akt2(+/+) mice. Urinary glucose excretion was significantly higher in akt2(-/-) mice compared with akt2(+/+) mice with or without fructose treatment. Moreover, the glucose-induced depolarization of proximal tubular cells was significantly smaller in isolated, perfused renal tubules from akt2(-/-) mice than in those from akt2(+/+) mice. In conclusion, Akt2/PKBß plays a role in the regulation of renal glucose transport.


Assuntos
Glucose/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Absorção , Animais , Feminino , Humanos , Camundongos , Camundongos Knockout , Modelos Animais , Oócitos/citologia , Oócitos/metabolismo , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Xenopus laevis
13.
Cell Physiol Biochem ; 25(2-3): 271-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20110688

RESUMO

Adenomatous polyposis coli (APC) is inactivated in familial adenomatous polyposis and sporadic colorectal cancer. Mice carrying defective APC (apc(Min/+)) spontaneously develop gastrointestinal tumors. APC binds GSK3beta, which phosphorylates beta-catenin thus fostering its degradation. beta-catenin upregulates the serum- and glucocorticoid-inducible kinase Sgk1, which inhibits GSK3beta. The present study explored the role of SGK1 in tumor growth of apc(Min/+)mice. apc(Min/+)mice were crossed with SGK1-knockout mice (sgk1(-/-)) and their wild type littermates (sgk1(+/+)) generating apc(Min/+)/sgk1(-/-)mice and apc(Min/+)/sgk1(+/+)mice. beta-catenin abundance was determined by Western blotting and confocal microscopy. As a result apc(Min/+)/sgk1(+/+)mice developed significantly more intestinal tumors than apc(Min/+)/sgk1(-/-)mice. Following chemical cancerogenesis, colonic beta-catenin protein abundance was significantly higher in sgk1(+/+)mice than in sgk1(-/-)mice. beta-catenin expression was significantly increased in HEK293 cells treated with dexamethasone for upregulation of Sgk1. In conclusion, SGK1 expression favors the development of intestinal tumors in APC-deficient mice, an effect at least partially due to enhanced beta-catenin protein abundance.


Assuntos
Proteína da Polipose Adenomatosa do Colo/deficiência , Proteínas Imediatamente Precoces/metabolismo , Neoplasias Intestinais/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Antineoplásicos Hormonais/farmacologia , Linhagem Celular , Dexametasona/farmacologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , beta Catenina/metabolismo
14.
IUBMB Life ; 61(7): 768-76, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19548318

RESUMO

The serum and glucocorticoid inducible kinase SGK1 was originally cloned from mammary tumor cells. SGK1 was found to be up-regulated in a variety of tumors, but down-regulated in several distinct tumors. Thus, evidence for a role of SGK1 in tumor growth remained conflicting. According to in vitro observations, SGK1 is up-regulated by the oncogene beta-catenin and negatively regulates the proapoptotic transcription factor FOXO3a, which in turn stimulates transcription of the Bcl2-interacting mediator BIM. This study aimed to define the role of SGK1 in colon carcinoma in vivo. SGK1 knockout mice (sgk1(-/-)) and their wild type littermates (sgk1(+/+)) were subjected to chemical cancerogenesis (intraperitoneal injection of 20 mg/kg 1,2-dimethylhydrazine followed by three cycles of 30 g/L synthetic dextran sulfate sodium for 7 days). Moreover, SGK1 was silenced in HEK293 cells. FOXO3a and BIM protein abundance was determined by Western blotting and immunohistochemistry. Following chemical cancerogenesis, sgk1(-/-)mice developed significantly less colonic tumors than sgk1(+/+)mice. According to Western blotting and immunohistochemistry, SGK1 deficiency enhanced the expression of FOXO3a and BIM both, in vitro and in vivo. SGK1 deficiency counteracts the development of colonic tumors, an effect at least in part due to up-regulation of FOXO3a and BIM.


Assuntos
Neoplasias do Colo/induzido quimicamente , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/fisiologia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/fisiologia , 1,2-Dimetilidrazina , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/biossíntese , Proteína 11 Semelhante a Bcl-2 , Hipóxia Celular , Neoplasias do Colo/patologia , Neoplasias do Colo/fisiopatologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/biossíntese , Regulação da Expressão Gênica , Humanos , Rim/embriologia , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/biossíntese , Células Tumorais Cultivadas
15.
Am J Physiol Regul Integr Comp Physiol ; 296(6): R1695-701, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19369585

RESUMO

The pore-forming K+-channel alpha-subunit KCNQ1 is expressed in a wide variety of tissues including heart, skeletal muscle, liver, and epithelia. Most recent evidence revealed an association of the KCNQ1 gene with the susceptibility to type 2 diabetes. KCNQ1 participates in the regulation of cell volume, which is, in turn, critically important for the regulation of metabolism by insulin. The present study explored the influence of KCNQ1 on insulin-induced cellular K+ uptake and glucose metabolism. Insulin (100 nM)-induced K+ uptake was determined in isolated perfused livers from KCNQ1-deficient mice (kcnq1(-/-)) and their wild-type littermates (kcnq1(+/+)). Moreover, plasma glucose and insulin levels, intraperitoneal glucose (3 g/kg) tolerance, insulin (0.15 U/kg)-induced hypoglycemia, and peripheral uptake of radiolabeled 3H-deoxy-glucose were determined in both genotypes. Insulin-stimulated hepatocellular K+ uptake was significantly more sustained in isolated perfused livers from kcnq1(-/-) mice than from kcnq1(+/+)mice. The decline of plasma glucose concentration following an intraperitoneal injection of insulin was again significantly more sustained in kcnq1(-/-) than in kcnq1(+/+) mice. Both fasted and nonfasted plasma glucose and insulin concentrations were significantly lower in kcnq1(-/-) than in kcnq1(+/+)mice. Following an intraperitoneal glucose injection, the peak plasma glucose concentration was significantly lower in kcnq1(-/-) than in kcnq1(+/+)mice. Uptake of 3H-deoxy-glucose into skeletal muscle, liver, kidney and lung tissue was significantly higher in kcnq1(-/-) than in kcnq1(+/+)mice. In conclusion, KCNQ1 counteracts the stimulation of cellular K+ uptake by insulin and thereby influences K+-dependent insulin signaling on glucose metabolism. The observations indicate that KCNQ1 is a novel molecule affecting insulin sensitivity of glucose metabolism.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Canal de Potássio KCNQ1/deficiência , Fígado/metabolismo , Potássio/metabolismo , Animais , Glicemia/metabolismo , Desoxiglucose/metabolismo , Feminino , Glucose/administração & dosagem , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 1/metabolismo , Injeções Intraperitoneais , Insulina/administração & dosagem , Insulina/sangue , Canal de Potássio KCNQ1/genética , Rim/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Transdução de Sinais , Fatores de Tempo
16.
Cell Physiol Biochem ; 23(1-3): 133-42, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19255508

RESUMO

Adenomatous polyposis coli (APC) is a tumor suppressor gene inactivated in familial adenomatous polyposis and sporadic colorectal cancer. Mice carrying a loss-of-function mutation in the apc gene (apc(Min/+)) spontaneously develop gastrointestinal tumors. APC fosters degradation of beta-catenin, which in turn upregulates the serum- and glucocorticoid-inducible kinase SGK1. SGK1 stimulates KCNQ1, which is required for luminal K+ recycling and thus for gastric acid secretion. BCECF-fluorescence was utilized to determine gastric acid secretion in isolated gastric glands from apc(Min/+) mice and their wild type littermates (apc(+/+)). Western blotting was employed to analyse beta-catenin and SGK1 expression and immunohistochemistry to determine KCNQ1 protein abundance. beta-catenin and SGK1 expression were enhanced in apc(Min/+) mice. Cytosolic pH was similar in apc(Min/+) mice and apc(+/+) mice. Na+-independent pH recovery following an ammonium pulse (DeltapH/min), which reflects H+/K+ ATPase activity, was, however, significantly faster in apc(Min/+) mice than in apc(+/+)mice. In both genotypes DeltapH/min was abolished in the presence of H+/K+ ATPase inhibitor omeprazole (100 microM). Treatment of apc(Min/+) and apc(+/+)mice with 5 microM forskolin 15 minutes prior to the experiment or increase in local K+-concentrations to 35 mM (replacing Na+/NMDG) significantly increased DeltapH/min and abrogated the differences between genotypes. The increase of DeltapH/min in apc(Min/+)mice required SGK1, as it was abolished by additional knockout of SGK1 (apc(Min/+)/sgk1(-/-)). In conclusion, basal gastric acid secretion is significantly enhanced in apc(Min/+)mice, pointing to a role of APC in the regulation of gastric acid secretion. The effect of APC requires H+/K+ ATPase activity and is at least partially due to SGK1-dependent upregulation of KCNQ1.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Western Blotting , Colforsina/farmacologia , Citometria de Fluxo , Imunofluorescência , Mucosa Gástrica/efeitos dos fármacos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Canal de Potássio KCNQ1/metabolismo , Camundongos , Camundongos Mutantes , Omeprazol/farmacologia , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , beta Catenina/metabolismo
17.
Pflugers Arch ; 458(3): 503-12, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19184092

RESUMO

Klotho, a membrane protein mainly expressed in parathyroid glands, kidney, and choroid plexus, counteracts aging and increases the life span. Accordingly, life span is significantly shorter in Klotho-deficient mice (klotho(-/-)) than in their wild-type littermates (klotho(+/+)). The pleotropic effects of Klotho include inhibition of 1,25-dihydroxyvitamin D(3)(1,25(OH)(2)D(3)) formation. Vitamin D-deficient diet reverses the shortening of life span in klotho(-/-) mice. In a variety of cells, 1,25(OH)(2)D(3) stimulates Ca(2+) entry. In erythrocytes, increased Ca(2+) entry stimulates suicidal erythrocyte death, which is characterized by cell shrinkage and phosphatidylserine exposure at the erythrocyte surface. The present study explored the putative impact of Klotho on eryptosis. According to Fluo3 fluorescence, cytosolic Ca(2+) concentration was significantly larger in klotho(-/-) erythrocytes as compared to klotho(+/+) erythrocytes. According to annexin V-binding, phosphatidylserine exposure was significantly enhanced, and according to forward scatter, cell volume significantly decreased in klotho(-/-) erythrocytes as compared to klotho(+/+) erythrocytes. Energy depletion (13 h glucose depletion) and oxidative stress (35 min 1 mM tert-butyl-hydroxyl-peroxide [tert-BOOH]) increased phosphatidylserine exposure to values again significantly larger in klotho(-/-) erythrocytes as compared to klotho(+/+) erythrocytes. Reticulocyte number was significantly increased in klotho (-/-) mice, pointing to enhanced erythrocyte turnover. Vitamin D-deficient diet reversed the enhanced Ca(2+) entry and annexin V-binding of klotho(-/-) erythrocytes. The present observations reveal a novel function of Klotho, i.e., the at least partially vitamin D-dependent regulation of cytosolic Ca(2+) activity in and suicidal death of erythrocytes.


Assuntos
Cálcio/metabolismo , Eritrócitos/citologia , Eritrócitos/fisiologia , Glucuronidase/metabolismo , Animais , Apoptose/fisiologia , Sinalização do Cálcio , Morte Celular/fisiologia , Células Cultivadas , Feminino , Proteínas Klotho , Masculino , Camundongos , Camundongos Knockout
18.
Am J Physiol Renal Physiol ; 296(4): F859-66, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158347

RESUMO

The hyperglycemia of diabetes mellitus increases the filtered glucose load beyond the maximal tubular transport rate and thus leads to glucosuria. Sustained hyperglycemia, however, may gradually increase the maximal renal tubular transport rate and thereby blunt the increase of urinary glucose excretion. The mechanisms accounting for the increase of renal tubular glucose transport have remained ill-defined. A candidate is the serum- and glucocorticoid-inducible kinase SGK1. The kinase has been shown to stimulate Na(+)-coupled glucose transport in vitro and mediate the stimulation of electrogenic intestinal glucose transport by glucocorticoids in vivo. SGK1 expression is confined to glomerula and distal nephron in intact kidneys but may extend to the proximal tubule in diabetic nephropathy. To explore whether SGK1 modifies glucose transport in diabetic kidneys, Akita mice (akita(+/-)), which develop spontaneous diabetes, have been crossbred with gene-targeted mice lacking SGK1 on one allele (sgk1(+/-)) to eventually generate either akita(+/-)/sgk1(-/-) or akita(+/-)/sgk1(+/+) mice. Both akita(+/-)/sgk1(-/-) and akita(+/-)/sgk1(+/+) mice developed profound hyperglycemia (>20 mM) within approximately 6 wk. Body weight and plasma glucose concentrations were not significantly different between these two genotypes. However, urinary excretion of glucose and urinary excretion of fluid, Na(+), and K(+), as well as plasma aldosterone concentrations, were significantly higher in akita(+/-)/sgk1(-/-) than in akita(+/-)/sgk1(+/+) mice. Studies in isolated perfused proximal tubules revealed that the electrogenic glucose transport was significantly lower in akita(+/-)/sgk1(-/-) than in akita(+/-)/sgk1(+/+) mice. The data provide the first evidence that SGK1 participates in the stimulation of renal tubular glucose transport in diabetic kidneys.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus/metabolismo , Nefropatias Diabéticas/metabolismo , Glicosúria/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Absorção , Fatores Etários , Aldosterona/sangue , Animais , Transporte Biológico , Peso Corporal , Creatinina/metabolismo , Diabetes Mellitus/sangue , Diabetes Mellitus/urina , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/urina , Modelos Animais de Doenças , Ingestão de Líquidos , Ingestão de Alimentos , Glicosúria/sangue , Glicosúria/urina , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Potenciais da Membrana , Camundongos , Camundongos Knockout , Potássio/sangue , Potássio/urina , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Sódio/sangue , Sódio/urina , Transportador 1 de Glucose-Sódio/metabolismo
19.
Cell Physiol Biochem ; 22(5-6): 725-34, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19088454

RESUMO

Pharmacological inhibition of phosphoinositide 3-kinase (PI3K) has previously been shown to enhance basal gastric acid secretion. Signaling of PI-3-kinase includes activation of the phosphoinositide dependent kinase PDK1. We thus hypothesized that PDK1 may influence gastric acid secretion. In the present study gastric acid secretion in mice expressing ñ20 % of PDK1 (pdk1(hm)) was compared to gastric acid secretion in their wild type littermates (pdk1(wt)). According to BCECF-fluorescence cytosolic pH in isolated gastric glands was similar in pdk1(hm) and in pdk1(wt) mice. Na(+)-independent pH recovery (DeltapH/min) following an ammonium pulse, which reflects K(+)/H(+)-ATPase activity, was however, significantly faster in pdk1(hm) than in pdk1(wt) mice. In both genotypes, DeltapH/min was abolished in the presence of K(+)/H(+)-ATPase inhibitor omeprazole (100 microM). Increase of local K(+)-concentrations to 35 mM (replacing Na(+)/NMDG) significantly increased DeltapH/min in both, pdk1(hm) and pdk1(wt) mice, and abrogated the differences between genotypes. Similarly, treatment with 5 microM forskolin as well as stimulation of protein kinase C with phorbolester phorbol 12 myristate 13 acetate (100 nM) enhanced DeltapH/min to almost identical values in pdk1(hm) and in pdk1(wt)mice. Protein kinase A inhibitor H89 (50 nM) decreased DeltapH/min in pdk1(hm) to values similar to those in pdk1(wt). In conclusion, deficient activity of PDK1 leads to a marked increase of gastric acid secretion. The present observations thus disclose a novel element in the regulation of gastric H(+) secretion.


Assuntos
Ácido Gástrico/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Carbacol/farmacologia , Colforsina/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Isoquinolinas/farmacologia , Camundongos , Omeprazol/farmacologia , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Potássio/farmacologia , Compostos de Amônio Quaternário/metabolismo , Sulfonamidas/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
20.
Cell Physiol Biochem ; 22(5-6): 735-44, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19088455

RESUMO

Pathological anxiety is paralleled by deficits in serotonergic and GABAergic neurotransmission in the amygdala. Conversely, anxiety disorders and depression may be reversed by brain-derived neurotrophic factor (BDNF). BDNF signaling involves Phosphatidylinositol 3-Kinase / 3-phosphoinositide-dependent protein kinase 1 (PI3K/PDK1). We thus hypothesized that impaired function of PDK1 might be associated with increased anxiety and concomitant neurotransmitter changes. Here we used the hypomorphic PDK1(hm) mouse to investigate anxiety behavior in different settings: PDK1(hm) mice differed from Wt littermates PDK1(WT) in several behavioral measures related to anxiety and exploration, namely in the open field, dark-light box, O-maze and startle response. Further we analyzed the brain substrate underlying this phenotype and found significantly decreased GABA, taurine and serotonin concentrations in the amygdala and olfactory bulb of PDK1(hm) mice, while BDNF and nerve growth factor (NGF) concentrations were not significantly different between PDK1(hm) and PDK1(WT) mice. These results suggest that impaired PI3K signaling in the PDK1(hm) mouse reduces concentrations of GABA and serotonin in anxiety related brain regions and can serve as a molecular substrate for behavior indicative for anxious and depressive-like mood states.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/enzimologia , Proteínas Serina-Treonina Quinases/deficiência , Serotonina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Tonsila do Cerebelo/anatomia & histologia , Animais , Ansiedade/fisiopatologia , Comportamento Animal , Peso Corporal , Ritmo Circadiano , Aprendizagem em Labirinto , Camundongos , Fatores de Crescimento Neural/metabolismo , Testes Neuropsicológicos , Neurotransmissores/metabolismo , Bulbo Olfatório/metabolismo , Tamanho do Órgão , Proteínas Serina-Treonina Quinases/metabolismo , Reflexo de Sobressalto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...