Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 6: 30925, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27499046

RESUMO

Putative functions of the heterotrimeric G-protein subunit Gαi2-dependent signaling include ion channel regulation, cell differentiation, proliferation and apoptosis. Erythrocytes may, similar to apoptosis of nucleated cells, undergo eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine (PS) exposure. Eryptosis may be triggered by increased cytosolic Ca(2+) activity and ceramide. In the present study, we show that Gαi2 is expressed in both murine and human erythrocytes and further examined the survival of erythrocytes drawn from Gαi2-deficient mice (Gαi2(-/-)) and corresponding wild-type mice (Gαi2(+/+)). Our data show that plasma erythropoietin levels, erythrocyte maturation markers, erythrocyte counts, hematocrit and hemoglobin concentration were similar in Gαi2(-/-) and Gαi2(+/+) mice but the mean corpuscular volume was significantly larger in Gαi2(-/-) mice. Spontaneous PS exposure of circulating Gαi2(-/-) erythrocytes was significantly lower than that of circulating Gαi2(+/+) erythrocytes. PS exposure was significantly lower in Gαi2(-/-) than in Gαi2(+/+) erythrocytes following ex vivo exposure to hyperosmotic shock, bacterial sphingomyelinase or C6 ceramide. Erythrocyte Gαi2 deficiency further attenuated hyperosmotic shock-induced increase of cytosolic Ca(2+) activity and cell shrinkage. Moreover, Gαi2(-/-) erythrocytes were more resistant to osmosensitive hemolysis as compared to Gαi2(+/+) erythrocytes. In conclusion, Gαi2 deficiency in erythrocytes confers partial protection against suicidal cell death.


Assuntos
Eriptose , Eritrócitos/citologia , Eritrócitos/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Animais , Sobrevivência Celular , Índices de Eritrócitos , Eritrócitos/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Humanos , Camundongos , Camundongos Knockout , Fosfatidilserinas/análise
2.
Curr Med Chem ; 23(6): 578-88, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26639094

RESUMO

Janus kinase-2 (JAK2) is a non-receptor tyrosine kinase signaling molecule that mediates the effects of various hormones and cytokines, including interferon, erythropoietin, leptin, and growth hormone. It also fosters tumor growth and modifies the activity of several nutrient transporters. JAK2 contributes to the regulation of the cell volume, protectS cells during energy depletion, proliferation, and aids the survival of tumor cells. Recently, JAK2 was identified as a powerful regulator of transport processes across the plasma membrane. Either directly or indirectly JAK2 may stimulate or inhibit transporter proteins, including ion channels, carriers and Na(+)/K(+) pumps. As a powerful regulator of transport mechanisms across the cell membrane, JAK2 regulates a wide variety of potassium, calcium, sodium and chloride ion channels, multiple Na+-coupled cellular carriers including EAAT1-4, NaPi-IIa, SGLT1, BoaT1, PepT1-2, CreaT1, SMIT1, and BGT1 as well as Na(+)/K(+)-ATPase. These cellular transport regulations contribute to various physiological and pathophysiological processes and thus exerting JAK2-sensitive effects. Future investigations will be important to determine whether JAK2 regulates cell-surface expression of other transporters and further elucidate underlying mechanisms governing JAK2 actions.


Assuntos
Proteínas de Transporte/metabolismo , Janus Quinase 2/metabolismo , Animais , Transporte Biológico , Humanos , Canais Iônicos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
3.
Kidney Blood Press Res ; 40(4): 403-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26227001

RESUMO

BACKGROUND/AIMS: Epidemiological evidence suggests that vitamin D deficiency is associated with anemia. The potent metabolite 1,25(OH)2 vitamin D3 [1,25(OH)2D3] activates various signaling cascades regulating a myriad of cellular functions including suicidal cell death or apoptosis. Suicidal death of erythrocytes or eryptosis is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine (PS) externalization. Stimulation of eryptosis may limit lifespan of circulating erythrocytes and thus cause anemia. In the present study, we explored the effect of a high vitamin D diet (10,000 I.U. vitamin D for 14 days) in mice on eryptosis. METHODS: Plasma concentrations of erythropoietin were estimated using an immunoassay kit, blood count using an electronic hematology particle counter, relative reticulocyte numbers using Retic-COUNT® reagent, PS exposure at the cell surface from annexin V binding, cell volume from forward scatter, and cytosolic Ca(2+) ([Ca(2+)]i) from Fluo3-fluorescence in FACS analysis. RESULTS: Vitamin D treatment decreased mean corpuscular volume, reticulocyte count, and plasma erythropoietin levels. Vitamin D treatment slightly but significantly decreased forward scatter but did not significantly modify spontaneous PS exposure and [Ca(2+)]i of freshly drawn erythrocytes. Vitamin D treatment augmented the stimulation of PS exposure and cell shrinkage following exposure to hyperosmotic shock (addition of 550 mM sucrose) or energy depletion (glucose removal) without significantly modifying [Ca(2+)]i. CONCLUSIONS: The present observations point to a subtle effect of exogenous vitamin D supplementation on erythrocyte survival.


Assuntos
Envelhecimento Eritrocítico/efeitos dos fármacos , Vitamina D/uso terapêutico , Vitaminas/uso terapêutico , Animais , Contagem de Células Sanguíneas , Cálcio/metabolismo , Tamanho Celular/efeitos dos fármacos , Dieta , Membrana Eritrocítica/efeitos dos fármacos , Eritropoetina/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Pressão Osmótica/efeitos dos fármacos , Fosfatidilserinas/sangue
4.
Am J Physiol Renal Physiol ; 305(12): F1645-55, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24107421

RESUMO

Cystinuria is an autosomal recessive disease caused by mutations in SLC3A1 (rBAT) and SLC7A9 (b(0,+)AT). Gene targeting of the catalytic subunit (Slc7a9) in mice leads to excessive excretion of cystine, lysine, arginine, and ornithine. Here, we studied this non-type I cystinuria mouse model using gene expression analysis, Western blotting, clearance, and brush-border membrane vesicle (BBMV) uptake experiments to further characterize the renal and intestinal consequences of losing Slc7a9 function. The electrogenic and BBMV flux studies in the intestine suggested that arginine and ornithine are transported via other routes apart from system b(0,+). No remarkable gene expression changes were observed in other amino acid transporters and the peptide transporters in the intestine and kidney. Furthermore, the glomerular filtration rate (GFR) was reduced by 30% in knockout animals compared with wild-type animals. The fractional excretion of arginine was increased as expected (∼100%), but fractional excretions of lysine (∼35%), ornithine (∼16%), and cystine (∼11%) were less affected. Loss of function of b(0,+)AT reduced transport of cystine and arginine in renal BBMVs and completely abolished the exchanger activity of dibasic amino acids with neutral amino acids. In conclusion, loss of Slc7a9 function decreases the GFR and increases the excretion of several amino acids to a lesser extent than expected with no clear regulation at the mRNA and protein level of alternative transporters and no increased renal epithelial uptake. These observations indicate that transporters located in distal segments of the kidney and/or metabolic pathways may partially compensate for Slc7a9 loss of function.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/deficiência , Diamino Aminoácidos/metabolismo , Cistina/metabolismo , Cistinúria/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Animais , Cistinúria/fisiopatologia , Modelos Animais de Doenças , Taxa de Filtração Glomerular/fisiologia , Rim/metabolismo , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Kidney Blood Press Res ; 37(1): 48-57, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23548792

RESUMO

BACKGROUND: Little is known about the effect of salt content of ingested fluid on intestinal transport processes. Osmosensitive genes include the serum- and glucocorticoid-inducible kinase SGK1, which is up-regulated by hyperosmolarity and cell shrinkage. SGK1 is in turn a powerful stimulator of the intestinal Na(+)/H(+) exchanger NHE3. The present study was thus performed to elucidate, whether the NaCl content of beverages influences NHE3 activity. METHODS: Mice were offered access to either plain water or isotonic saline ad libitum. NHE3 transcript levels and protein abundance in intestinal tissue were determined by confocal immunofluorescent microscopy, RT-PCR and western blotting, cytosolic pH (pHi) in intestinal cells from 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) fluorescence and Na(+)/H(+) exchanger activity from the Na(+) dependent realkalinization following an ammonium pulse. RESULTS: Saline drinking significantly enhanced fluid intake and increased NHE3 transcript levels, NHE3 protein and Na(+)/H(+) exchanger activity. CONCLUSIONS: Salt content of ingested fluid has a profound effect on intestinal Na(+)/H(+) exchanger expression and activity.


Assuntos
Ingestão de Alimentos/fisiologia , Jejuno/metabolismo , Cloreto de Sódio/administração & dosagem , Trocadores de Sódio-Hidrogênio/biossíntese , Regulação para Cima/fisiologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Jejuno/efeitos dos fármacos , Masculino , Camundongos , Técnicas de Cultura de Órgãos , Trocador 3 de Sódio-Hidrogênio , Regulação para Cima/efeitos dos fármacos
6.
Am J Physiol Gastrointest Liver Physiol ; 303(11): G1212-9, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23019198

RESUMO

The oxidative stress responsive kinase 1 (OSR1) contributes to WNK (with no K)-dependent regulation of renal tubular salt transport, renal salt excretion, and blood pressure. Little is known, however, about a role of OSR1 in the regulation of intestinal salt transport. The present study thus explored whether OSR1 is expressed in intestinal tissue and whether small intestinal Na(+)/H(+) exchanger (NHE), small intestinal Na(+)-glucose cotransport (SGLT1), and/or colonic epithelium Na(+) channel (ENaC) differ between knockin mice carrying one allele of WNK-resistant OSR1 (osr1(+/KI)) and wild-type mice (osr1(+/+)). OSR1 protein abundance was determined by Western blotting, cytosolic pH from BCECF fluorescence, NHE activity from Na(+)-dependent realkalinization following an ammonium pulse, SGLT1 activity from glucose-induced current, and colonic ENaC activity from amiloride-sensitive transepithelial current in Ussing chamber experiments. As a result, OSR1 protein was expressed in small intestine of both osr1(+/KI) mice and osr1(+/+) mice. Daily fecal Na(+), K(+), and H(2)O excretion and jejunal SGLT1 activity were lower, whereas small intestinal NHE activity and colonic ENaC activity were higher in osr1(+/KI) mice than in osr1(+/+) mice. NHE3 inhibitor S-3226 significantly reduced NHE activity in both genotypes but did not abrogate the difference between the genotypes. Plasma osmolarity, serum antidiuretic hormone, plasma aldosterone, and plasma corticosterone concentrations were similar in both genotypes. Small intestinal NHE3 and colonic α-ENaC protein abundance were not significantly different between genotypes, but colonic phospho-ß-ENaC (ser633) was significantly higher in osr1(+/KI) mice. In conclusion, OSR1 is expressed in intestine and partial WNK insensitivity of OSR1 increases intestinal NHE activity and colonic ENaC activity.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Animais , Canais Epiteliais de Sódio/metabolismo , Técnicas de Introdução de Genes , Guanidinas/farmacologia , Metacrilatos/farmacologia , Camundongos , Transportador 1 de Glucose-Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo
7.
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
8.
Endocrinology ; 153(4): 1783-94, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22294744

RESUMO

Glucocorticoid (GC) treatment of inflammatory disorders, such as inflammatory bowel disease, causes deranged metabolism, in part by enhanced intestinal resorption of glucose. However, the underlying molecular mechanism is poorly understood. Hence, we investigated transcriptional control of genes reported to be involved in glucose uptake in the small intestine after GC treatment and determined effects of GC on electrogenic glucose transport from transepithelial currents. GR(villinCre) mice lacking the GC receptor (GR) in enterocytes served to identify the target cell of GC treatment and the requirement of the GR itself; GR(dim) mice impaired in dimerization and DNA binding of the GR were used to determine the underlying molecular mechanism. Our findings revealed that oral administration of dexamethasone to wild-type mice for 3 d increased mRNA expression of serum- and GC-inducible kinase 1, sodium-coupled glucose transporter 1, and Na(+)/H(+) exchanger 3, as well as electrogenic glucose transport in the small intestine. In contrast, GR(villinCre) mice did not respond to GC treatment, neither with regard to gene activation nor to glucose transport. GR(dim) mice were also refractory to GC, because dexamethasone treatment failed to increase both, gene expression and electrogenic glucose transport. In addition, the rise in blood glucose levels normally observed after GC administration was attenuated in both mutant mouse strains. We conclude that enhanced glucose transport in vivo primarily depends on gene regulation by the dimerized GR in enterocytes, and that this mechanism contributes to GC-induced hyperglycemia.


Assuntos
Dexametasona/farmacologia , Dimerização , Enterócitos/metabolismo , Glucocorticoides/farmacologia , Glucose/metabolismo , Absorção Intestinal/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Enterócitos/citologia , Enterócitos/efeitos dos fármacos , Feminino , Proteínas Imediatamente Precoces/metabolismo , Interleucina-6/metabolismo , Jejuno/citologia , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Modelos Animais , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Glucocorticoides/deficiência , Receptores de Glucocorticoides/genética , Transportador 1 de Glucose-Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
Diabetes ; 61(1): 187-96, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22124465

RESUMO

To clarify the physiological role of Na(+)-D-glucose cotransporter SGLT1 in small intestine and kidney, Sglt1(-/-) mice were generated and characterized phenotypically. After gavage of d-glucose, small intestinal glucose absorption across the brush-border membrane (BBM) via SGLT1 and GLUT2 were analyzed. Glucose-induced secretion of insulinotropic hormone (GIP) and glucagon-like peptide 1 (GLP-1) in wild-type and Sglt1(-/-) mice were compared. The impact of SGLT1 on renal glucose handling was investigated by micropuncture studies. It was observed that Sglt1(-/-) mice developed a glucose-galactose malabsorption syndrome but thrive normally when fed a glucose-galactose-free diet. In wild-type mice, passage of D-glucose across the intestinal BBM was predominantly mediated by SGLT1, independent the glucose load. High glucose concentrations increased the amounts of SGLT1 and GLUT2 in the BBM, and SGLT1 was required for upregulation of GLUT2. SGLT1 was located in luminal membranes of cells immunopositive for GIP and GLP-1, and Sglt1(-/-) mice exhibited reduced glucose-triggered GIP and GLP-1 levels. In the kidney, SGLT1 reabsorbed ∼3% of the filtered glucose under normoglycemic conditions. The data indicate that SGLT1 is 1) pivotal for intestinal mass absorption of d-glucose, 2) triggers the glucose-induced secretion of GIP and GLP-1, and 3) triggers the upregulation of GLUT2.


Assuntos
Glucose/farmacocinética , Incretinas/metabolismo , Absorção Intestinal/genética , Transportador 1 de Glucose-Sódio/fisiologia , Animais , Feminino , Glucose/farmacologia , Glicosúria/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Intestino Delgado/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo
10.
Pflugers Arch ; 462(3): 489-94, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21735060

RESUMO

The serum- and glucocorticoid-inducible kinase SGK1 has previously been shown to mediate the glucocorticoid-dependent stimulation of several intestinal transport systems including the electrogenic glucose transporter SGLT1. In squamous carcinoma cells, SGK1 expression is stimulated by 1,25(OH)2D3, the biologically active metabolite of vitamin D. The present study explored whether vitamin D influences the intestinal SGLT1 activity. Jejunal SGLT1 activity was determined by Ussing chamber experiments. Under a normal diet, the electrogenic glucose transport was similar in SGK1 knockout (sgk1 ( -/- )) and wild type mice (sgk1 ( +/+ )). Following a vitamin D-rich diet (14 days 10,000 I.U. vitamin D), the SGK1 transcript levels as well as the SGLT1 protein abundance were increased in sgk1(+/+) mice. Moreover, SGLT1 activity was increased in sgk1(+/+) mice but not in sgk1(-/-) mice following a vitamin D-rich diet. Furthermore, an oral glucose load was followed by an increase in the plasma glucose concentration to significantly higher values in sgk1(+/+) mice treated with a vitamin D-rich diet than in untreated sgk1(+/+) mice. In conclusion, vitamin D treatment upregulates the expression of SGK1, which in turn enhances SGLT1 activity.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Vitamina D/administração & dosagem , Animais , Células CACO-2 , Dieta , Glucose/metabolismo , Teste de Tolerância a Glucose , Humanos , Proteínas Imediatamente Precoces/genética , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Transportador 1 de Glucose-Sódio/genética
11.
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
12.
Cell Physiol Biochem ; 26(4-5): 641-6, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21063101

RESUMO

Glycogen synthase kinase 3 GSK3ß participates in a wide variety of functions including regulation of glucose metabolism. It is ubiquitously expressed including epithelial tissues. However, whether GSK3ß participates in the regulation of epithelial transport is not known. The present study thus explored whether GSK3ß influences the Na(+)-coupled transport of glucose. To this end, SGLT1 was expressed in Xenopus oocytes with or without GSK3ß and glucose-induced current (I(g)) determined by dual electrode voltage clamp. In Xenopus oocytes expressing SGLT1 but not in water-injected oocytes glucose induced an inwardly directed I(g), which was significantly enhanced by coexpression of GSK3ß. According to chemiluminescence and confocal microscopy, GSK3ß increased the SGLT1 protein abundance in the oocyte cell membrane. To explore whether GSK3ß sensitivity of SGLT1 participates in the regulation of electrogenic intestinal glucose transport, Ussing chamber experiments were performed in intestinal segments from gene-targeted knockin mice with mutated and thus PKB/SGK-resistant GSK3α,ß (gsk3(KI)), in which the serine of the PKB/SGK phosphorylation site was replaced by alanine, and from wild type mice (gsk3(WT)). The glucose-induced current was significantly larger in gsk3(KI) than in gsk3(WT) mice. The present observations reveal a novel function of GSK3, i.e. the stimulation of Na(+)-coupled glucose transport.


Assuntos
Glucose/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Feminino , Técnicas de Introdução de Genes , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Masculino , Camundongos , Mutação , Oócitos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo , Xenopus/crescimento & desenvolvimento
13.
Cell Physiol Biochem ; 26(4-5): 699-706, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21063107

RESUMO

The function of mast cells is modified by the phosphoinositol-3 (PI3)-kinase pathway. The kinase signals partially through the phosphoinositide-dependent kinase PDK1, which on the one hand activates the serum- and glucocorticoid- inducible kinase SGK1 and on the other hand activates protein kinase PKCδ. SGK1 participates in the stimulation of Ca(2+) entry and degranulation, PKCδ inhibits degranulation. The present experiments explored the role of PDK1 in mast cell function. As mice completely lacking PDK1 are not viable, experiments have been performed in mast cells isolated from bone marrow (BMMCs) of PDK1 hypomorphic mice (pdk1(hm)) and their wild-type littermates (pdk1(wt)). Antigen stimulation via the FceRI receptor was followed by Ca(2+) entry leading to increase of cytosolic Ca(2+) activity in pdk1(wt) BMMCs, an effect significantly blunted in pdk1(hm) BMMCs. In contrast, Ca(2+) release from intracellular stores was not different between BMMCs of the two genotypes. The currents through Ca(2+)-activated K(+) channels following antigen exposure were again significantly larger in pdk1(wt) than in pdk1(hm) cells. The Ca(2+) ionophore ionomycin (1 µM) increased the K(+) channel conductance to similar values in both genotypes. ß-hexosaminidase and histamine release were similar in pdk1(wt) BMMCs and pdk1(hm) BMMCs. PKCδ inhibitor rottlerin increased ß-hexosaminidase release in pdk1(wt) BMMCs but not in pdk1(hm) BMMCs. Phosphorylation of PKCδ and of the SGK1 target NDRG1, was stimulated by the antigen in pdk1(wt) but not in pdk1(hm) cells. The observations reveal a role for PDK1 in the regulation of Ca(2+) entry into and degranulation of murine mast cells.


Assuntos
Cálcio/metabolismo , Mastócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Acetofenonas/farmacologia , Animais , Benzopiranos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Histamina/metabolismo , Proteínas Imediatamente Precoces , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ionomicina/farmacologia , Ionóforos/farmacologia , Mastócitos/efeitos dos fármacos , Mastócitos/enzimologia , Camundongos , Técnicas de Patch-Clamp , Fosforilação , Canais de Potássio/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Proteínas Serina-Treonina Quinases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
14.
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
15.
Mol Nutr Food Res ; 54(12): 1833-41, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20603831

RESUMO

SCOPE: Thymol is a component of several plants with antimicrobial activity. Little is known about the effects of thymol on immune cells of the host. This study addressed the effects of thymol on dendritic cells (DCs), regulators of innate and adaptive immunity. METHODS AND RESULTS: Immunohistochemistry, Western blotting and fluorescence-activated cell sorting analysis were performed in bone marrow-derived DCs either from wild-type mice or from mice lacking acid sphingomyelinase (ASM⁻/⁻) treated and untreated for 24 h with thymol (2-100 µg/mL). Thymol treatment resulted in activation of ASM, stimulation of ceramide formation, downregulation of anti-apoptotic Bcl-2 and Bcl-xL proteins, activation of caspase 3 and caspase 8, DNA fragmentation as well as cell membrane scrambling. The effects were dependent on the presence of ASM and were lacking in ASM⁻/⁻ mice or in wild-type DCs treated with sphingomyelinase inhibitor amitriptyline. CONCLUSION: Thymol triggers suicidal DC death, an effect mediated by and requiring activation of ASM.


Assuntos
Morte Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Esfingomielina Fosfodiesterase/metabolismo , Timol/farmacologia , Animais , Apoptose , Caspase 3/metabolismo , Caspase 8/metabolismo , Células Cultivadas , Ceramidas/biossíntese , Fragmentação do DNA , Células Dendríticas/enzimologia , Regulação para Baixo , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo
16.
Cell Physiol Biochem ; 25(6): 715-22, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20511717

RESUMO

The phosphoinositol 3 kinase (PI3K) and the phosphoinositide dependent kinase (PDK1) stimulate the serum and glucocorticoid inducible kinase (SGK) and protein kinase B (PKB/Akt) isoforms, kinases stimulating a variety of transporters. Most recently, SGK1 was shown to stimulate the peptide transporters PepT1 and PepT2, and to mediate the glucocorticoid stimulation of PepT1. Basal electrogenic intestinal peptide transport was, however, not dependent on the presence of SGK1. The present study explored whether basal electrogenic intestinal peptide transport is dependent on PI3K or PDK1. To this end, peptide transport in intestinal segments was determined utilizing Ussing chamber analysis. Cytosolic pH (pH(i)) was determined by BCECF fluorescence. The luminal addition of 5 mM dipeptide gly-gly induced a current (Ip) across intestinal segments. Ip was significantly decreased in the presence of PI3 kinase inhibitors Wortmannin (1 microM) or LY294002 (50 microM). Exposure of isolated intestinal cells to 5 mM gly-gly was followed by cytosolic acidification (DeltapH(i)), which was significantly blunted by Wortmannin and by LY294002. Both, Ip and DeltapHi were significantly smaller in PDK1 hypomorphic mice (pdk(1flfl)) than in their wild type littermates (pdk1(wt)). In conclusion, PI3K and PDK1 participate in the regulation of basal peptide transport.


Assuntos
Dipeptídeos/metabolismo , Mucosa Intestinal/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Intestinos/citologia , Camundongos
17.
Cell Physiol Biochem ; 25(2-3): 203-10, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20110681

RESUMO

Intestinal Na(+)-coupled glucose transporter SGLT1 determines the rate of glucose transport, which in turn influences glucose-induced insulin release and development of obesity. The present study explored effects of Gum Arabic (GA), a dietary polysaccharide from dried exudates of Acacia Senegal, on intestinal glucose transport and body weight in wild-type C57Bl/6 mice. Treatment with GA (100 g/l) in drinking water for four weeks did not affect intestinal SGLT1 transcript levels but decreased SGLT1 protein abundance in jejunal brush border membrane vesicles. Glucose-induced jejunal short-circuit currents revealed that GA treatment decreased electrogenic glucose transport. Drinking a 20% glucose solution for four weeks significantly increased body weight and fasting plasma glucose concentrations, effects significantly blunted by simultaneous treatment with GA. GA further significantly blunted the increase in body weight, fasting plasma glucose and fasting insulin concentrations during high fat diet. In conclusion, the present observations disclose a completely novel effect of gum arabic, i.e. its ability to decrease intestinal SGLT1 expression and activity and thus to counteract glucose-induced obesity.


Assuntos
Goma Arábica/farmacologia , Jejuno/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Animais , Regulação para Baixo , Glucose/metabolismo , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Transportador 1 de Glucose-Sódio/genética
18.
Pflugers Arch ; 459(1): 191-202, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19672619

RESUMO

According to recent in vitro experiments, the peptide transporter PepT2 is stimulated by the serum- and glucocorticoid-inducible kinase SGK1. The present study explored the contribution of SGK1 to the regulation of electrogenic intestinal peptide transport. Intestinal PepT1 was expressed in Xenopus oocytes, and peptide transport was determined by dual electrode voltage clamping. Peptide transport in intestinal segments was determined utilizing Ussing chamber. Cytosolic pH (pH( i )) was determined by BCECF fluorescence and Na(+)/H(+) exchanger activity was estimated from Na(+)-dependent pH recovery (pH ( i )) following an ammonium pulse. In PepT1-expressing Xenopus oocytes, coexpression of SGK1 enhanced electrogenic peptide transport. Intestinal transport and pH( i ) of untreated mice were similar in SGK1 knockout mice (sgk1 ( -/- )) and their wild-type littermates (sgk1 ( +/+ )). Glucocorticoid treatment (4 days 10 microg/g body weight (bw)/day dexamethasone) increased peptide transport in sgk1 ( +/+ ) but not in sgk1 (-/-) mice. Irrespective of dexamethasone treatment, luminal peptide (5 mM glycyl-glycine) led to a similar early decrease of pH( i ) in sgk1 (-/-) and sgk1 (+/+) mice, but to a more profound and sustained decline of pH( i ) in sgk1 (-/-) than in sgk1 ( +/+ ) mice. In the presence and absence of glycyl-glycine, pH ( i ) was significantly enhanced by dexamethasone treatment in sgk1 ( +/+ ) mice, an effect significantly blunted in sgk1 ( -/- ) mice. During sustained exposure to glycyl-glycine, pH ( i ) was significantly larger in sgk1 (+/+) mice than in sgk1 (-/-) mice, irrespective of dexamethasone treatment. In conclusion, basal intestinal peptide transport does not require stimulation by SGK1. Glucocorticoid treatment stimulates both Na(+)/H(+) exchanger activity and peptide transport, effects partially dependent on SGK1. Moreover, chronic exposure to glycyl-glycine stimulates Na(+)/H(+) exchanger activity, an effect again involving SGK1.


Assuntos
Dexametasona/metabolismo , Glucocorticoides/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Simportadores/metabolismo , Animais , Dipeptídeos/metabolismo , Imunofluorescência , Glicilglicina/metabolismo , Concentração de Íons de Hidrogênio , Intestinos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trocadores de Sódio-Hidrogênio/metabolismo , Xenopus
19.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R571-5, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19494170

RESUMO

Adenomatous polyposis coli (APC) fosters degradation of beta-catenin, a multifunctional protein upregulating the serum- and glucocorticoid-inducible kinase (SGK1). SGK1 regulates a wide variety of renal transport processes. The present study explored the possibility that APC influences renal function. To this end, metabolic cage experiments were performed in mice carrying a loss-of-function mutation in the APC gene (apc(Min/+)), their wild-type littermates (apc(+/+)), and apc(Min/+) mice lacking functional SGK1 (apc(Min/+)/sgk1(-/-)). As a result, mean body weight, food intake, fluid intake, salt appetite, urinary flow, as well as plasma Na(+) and K(+) concentrations were similar in apc(Min/+) mice, apc(+/+) mice, and apc(Min/+)/sgk1(-/-) mice. Glomerular filtration rate and absolute renal Na(+) excretion were decreased, and fractional urinary K(+) excretion was enhanced in apc(Min/+) mice. The antinatriuresis, but not the hypofiltration and kaliuresis was partially reversed by additional lack of SGK1. Plasma corticosterone and aldosterone concentrations were significantly enhanced in apc(Min/+) mice. While the plasma corticosterone concentration was similar in apc(+/+) mice and apc(Min/+)/sgk1(-/-) mice, plasma aldosterone was even higher in apc(Min/+)/sgk1(-/-) mice than in apc(Min/+) mice. The hyperaldosteronism of apc(Min/+) mice was paralleled by significantly elevated plasma volume and blood pressure. The experiments reveal an influence of defective APC on adrenal hormone release and renal function, effects partially but not completely explained by APC dependence of SGK1 expression.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Pressão Sanguínea , Volume Sanguíneo , Hiperaldosteronismo/fisiopatologia , Hipertensão/fisiopatologia , Rim/fisiopatologia , Mutação , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Aldosterona/sangue , Animais , Pressão Sanguínea/genética , Volume Sanguíneo/genética , Peso Corporal , Corticosterona/sangue , Ingestão de Líquidos , Ingestão de Alimentos , Taxa de Filtração Glomerular , Homeostase , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Natriurese , Potássio/sangue , Potássio/urina , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Sódio/sangue , Sódio/urina , Cloreto de Sódio na Dieta/administração & dosagem , Urodinâmica , Proteínas Wnt/metabolismo
20.
Kidney Blood Press Res ; 32(2): 119-27, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19401625

RESUMO

BACKGROUND/AIMS: Serum- and glucocorticoid-inducible kinase 1 (SGK1) stimulates the epithelial sodium channel (ENaC), renal outer medullary K(+) channel 1, Na(+)/K(+)-ATPase and presumably the Na(+)-Cl(-) cotransporter (NCC). SGK1-deficient mice (sgk(-/-)) show a compensated salt-losing phenotype with secondary hyperaldosteronism. The present experiments explored the role of SGK1 in the response to diuretics. METHODS: sgk1(-/-) mice and their wild-type littermates (sgk1(+/+)) were treated with the ENaC blocker triamterene (200 mg/l), the Na(+)-K(+)-2Cl(-) cotransport inhibitor furosemide (125 mg/l), the NCC blocker hydrochlorothiazide (400 mg/l) and the mineralocorticoid receptor blocker canrenoate (800 mg/l) for 8 days. Renal SGK1 expression was studied using quantitative RT-PCR and immunofluorescence. RESULTS: Diuretic treatment increased SGK1 mRNA and protein expression in the kidney of wild-type sgk1(+/+) mice. The responses to furosemide, hydrochlorothiazide or canrenoate were not different between sgk1(+/+) and sgk1(-/-) mice, and were accompanied by moderate increases in plasma aldosterone and urea concentrations. However, treatment with triamterene in sgk1(-/-) mice (but not in sgk1(+/+) mice) led to severe, eventually lethal, body weight loss as well as increases in plasma aldosterone, urea and K(+) concentrations. CONCLUSIONS: SGK1 is required for diuretic tolerance to triamterene. The observations confirm the impaired kaliuretic potency of sgk1(-/-) mice and point to a role of SGK1 in renal Na(+) reabsorption by mechanisms other than ENaC.


Assuntos
Diuréticos/farmacologia , Marcação de Genes/métodos , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Animais , Tolerância a Medicamentos , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Canais Epiteliais de Sódio/fisiologia , Proteínas Imediatamente Precoces/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases/sangue , Triantereno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA