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.
Kidney Int ; 93(5): 1131-1141, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29395333

RESUMO

It is accepted that osteoblasts/osteocytes are the major source for circulating fibroblast growth factor 23 (FGF23). However, erythropoietic cells of bone marrow also express FGF23. The modulation of FGF23 expression in bone marrow and potential contribution to circulating FGF23 has not been well studied. Moreover, recent studies show that plasma FGF23 may increase early during acute kidney injury (AKI). Erythropoietin, a kidney-derived hormone that targets erythropoietic cells, increases in AKI. Here we tested whether an acute increase of plasma erythropoietin induces FGF23 expression in erythropoietic cells of bone marrow thereby contributing to the increase of circulating FGF23 in AKI. We found that erythroid progenitor cells of bone marrow express FGF23. Erythropoietin increased FGF23 expression in vivo and in bone marrow cell cultures via the homodimeric erythropoietin receptor. In experimental AKI secondary to hemorrhagic shock or sepsis in rodents, there was a rapid increase of plasma erythropoietin, and an induction of bone marrow FGF23 expression together with a rapid increase of circulating FGF23. Blockade of the erythropoietin receptor fully prevented the induction of bone marrow FGF23 and partially suppressed the increase of circulating FGF23. Finally, there was an early increase of both circulating FGF23 and erythropoietin in a cohort of patients with severe sepsis who developed AKI within 48 hours of admission. Thus, increases in plasma erythropoietin and erythropoietin receptor activation are mechanisms implicated in the increase of plasma FGF23 in AKI.


Assuntos
Injúria Renal Aguda/sangue , Células da Medula Óssea/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoetina/sangue , Fatores de Crescimento de Fibroblastos/sangue , Injúria Renal Aguda/etiologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Modelos Animais de Doenças , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Fator de Crescimento de Fibroblastos 23 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Estudos Prospectivos , Ratos Sprague-Dawley , Receptores da Eritropoetina/agonistas , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes/farmacologia , Sepse/sangue , Sepse/complicações , Choque Hemorrágico/sangue , Choque Hemorrágico/complicações , Fatores de Tempo , Regulação para Cima
2.
Channels (Austin) ; 11(5): 388-398, 2017 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-28636485

RESUMO

Renal sodium reabsorption depends on the activity of the Na+,K+-ATPase α/ß heterodimer. Four α (α1-4) and 3 ß (ß1-3) subunit isoforms have been described. It is accepted that renal tubule cells express α1/ß1 dimers. Aldosterone stimulates Na+,K+-ATPase activity and may modulate α1/ß1 expression. However, some studies suggest the presence of ß3 in the kidney. We hypothesized that the ß3 isoform of the Na+,K+-ATPase is expressed in tubular cells of the distal nephron, and modulated by mineralocorticoids. We found that ß3 is highly expressed in collecting duct of rodents, and that mineralocorticoids decreased the expression of ß3. Thus, we describe a novel molecular mechanism of sodium pump modulation that may contribute to the effects of mineralocorticoids on sodium reabsorption.


Assuntos
Túbulos Renais/metabolismo , Mineralocorticoides/farmacologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Aldosterona/farmacologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Agonistas do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/metabolismo , Masculino , Ratos Sprague-Dawley
3.
Am J Physiol Renal Physiol ; 310(11): F1216-28, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26911844

RESUMO

Extracellular proton-secreting transport systems that contribute to extracellular pH include the vacuolar H(+)-ATPase (V-ATPase). This pump, which mediates ATP-driven transport of H(+) across membranes, is involved in metastasis. We previously showed (Alzamora R, Thali RF, Gong F, Smolak C, Li H, Baty CJ, Bertrand CA, Auchli Y, Brunisholz RA, Neumann D, Hallows KR, Pastor-Soler NM. J Biol Chem 285: 24676-24685, 2010) that V-ATPase A subunit phosphorylation at Ser-175 is important for PKA-induced V-ATPase activity at the membrane of kidney intercalated cells. However, Ser-175 is also located within a larger phosphorylation consensus sequence for Aurora kinases, which are known to phosphorylate proteins that contribute to the pathogenesis of metastatic carcinomas. We thus hypothesized that Aurora kinase A (AURKA), overexpressed in aggressive carcinomas, regulates the V-ATPase in human kidney carcinoma cells (Caki-2) via Ser-175 phosphorylation. We found that AURKA is abnormally expressed in Caki-2 cells, where it binds the V-ATPase A subunit in an AURKA phosphorylation-dependent manner. Treatment with the AURKA activator anacardic acid increased V-ATPase expression and activity at the plasma membrane of Caki-2 cells. In addition, AURKA phosphorylates the V-ATPase A subunit at Ser-175 in vitro and in Caki-2 cells. Immunolabeling revealed that anacardic acid induced marked membrane accumulation of the V-ATPase A subunit in transfected Caki-2 cells. However, anacardic acid failed to induce membrane accumulation of a phosphorylation-deficient Ser-175-to-Ala (S175A) A subunit mutant. Finally, S175A-expressing cells had decreased migration in a wound-healing assay compared with cells expressing wild-type or a phospho-mimetic Ser-175-to-Asp (S175D) mutant A subunit. We conclude that AURKA activates the V-ATPase in kidney carcinoma cells via phosphorylation of Ser-175 in the V-ATPase A subunit. This regulation contributes to kidney carcinoma V-ATPase-mediated extracellular acidification and cell migration.


Assuntos
Aurora Quinase A/metabolismo , Carcinoma/metabolismo , Neoplasias Renais/metabolismo , Rim/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Ácidos Anacárdicos/farmacologia , Carcinoma/patologia , Linhagem Celular Tumoral , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Neoplasias Renais/patologia , Fosforilação/efeitos dos fármacos
4.
J Physiol ; 589(Pt 21): 5091-107, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21911611

RESUMO

The KCNQ1 potassium channel associates with various KCNE ancillary subunits that drastically affect channel gating and pharmacology. Co-assembly with KCNE3 produces a current with nearly instantaneous activation, some time-dependent activation at very positive potentials, a linear current-voltage relationship and a 10-fold higher sensitivity to chromanol 293B. KCNQ1:KCNE3 channels are expressed in colonic crypts and mediate basolateral K(+) recycling required for Cl(-) secretion. We have previously reported the female-specific anti-secretory effects of oestrogen via KCNQ1:KCNE3 channel inhibition in colonic crypts. This study was designed to determine whether sex and oestrogen regulate the expression and function of KCNQ1 and KCNE3 in rat distal colon. Colonic crypts were isolated from Sprague-Dawley rats and used for whole-cell patch-clamp and to extract total RNA and protein. Sheets of epithelium were used for short-circuit current recordings. KCNE1 and KCNE3 mRNA and protein abundance were significantly higher in male than female crypts. No expression of KCNE2 was found and no difference was observed in KCNQ1 expression between male and female (at oestrus) colonic crypts. Male crypts showed a 2.2-fold higher level of association of KCNQ1 and KCNE3 compared to female cells. In female colonic crypts, KCNQ1 and KCNE3 protein expression fluctuated throughout the oestrous cycle and 17ß-oestradiol (E2 10 nM) produced a rapid (<15 min) dissociation of KCNQ1 and KCNE3 in female crypts only. Whole-cell K(+) currents showed a linear current-voltage relationship in male crypts, while K(+) currents in colonic crypts isolated from females displayed voltage-dependent outward rectification. Currents in isolated male crypts and epithelial sheets were 10-fold more sensitive to specific KCNQ1 inhibitors, such as chromanol 293B and HMR-1556, than in female. The effect of E2 on K(+) currents mediated by KCNQ1 with or without different ß-subunits was assayed from current-voltage relations elicited in CHO cells transfected with KCNQ1 and KCNE3 or KCNE1 cDNA. E2 (100 nM) reduced the currents mediated by the KCNQ1:KCNE3 potassium channel and had no effect on currents via KCNQ1:KCNE1 or KCNQ1 alone. Currents mediated by the complex formed by KCNQ1 and the mutant KCNE3-S82A ß-subunit (mutation of the site for PKCδ-promoted phosphorylation and modulation of the activity of KCNE3) showed rapid run-down and insensitivity to E2. Together, these data suggest that oestrogen regulates the expression of the KCNE1 and KCNE3 and with it the gating and pharmacological properties of the K(+) conductance required for Cl(-) secretion. The decreased association of the KCNQ1:KCNE3 channel complex promoted by oestrogen exposure underlies the molecular mechanism for the sexual dimorphism and oestrous cycle dependence of the anti-secretory actions of oestrogen in the intestine.


Assuntos
Colo/fisiologia , Estrogênios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Feminino , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Canal de Potássio KCNQ1/fisiologia , Masculino , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Caracteres Sexuais
5.
Front Physiol ; 2: 33, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21747769

RESUMO

Berberine is a plant alkaloid with multiple pharmacological actions, including antidiarrhoeal activity and has been shown to inhibit Cl(-) secretion in distal colon. The aims of this study were to determine the molecular signaling mechanisms of action of berberine on Cl(-) secretion and the ion transporter targets. Monolayers of T84 human colonic carcinoma cells grown in permeable supports were placed in Ussing chambers and short-circuit current measured in response to secretagogues and berberine. Whole-cell current recordings were performed in T84 cells using the patch-clamp technique. Berberine decreased forskolin-induced short-circuit current in a concentration-dependent manner (IC(50) 80 ± 8 µM). In apically permeabilized monolayers and whole-cell current recordings, berberine inhibited a cAMP-dependent and chromanol 293B-sensitive basolateral membrane K(+) current by 88%, suggesting inhibition of KCNQ1 K(+) channels. Berberine did not affect either apical Cl(-) conductance or basolateral Na(+)-K(+)-ATPase activity. Berberine stimulated p38 MAPK, PKCα and PKA, but had no effect on p42/p44 MAPK and PKCδ. However, berberine pre-treatment prevented stimulation of p42/p44 MAPK by epidermal growth factor. The inhibitory effect of berberine on Cl(-) secretion was partially blocked by HBDDE (∼65%), an inhibitor of PKCα and to a smaller extent by inhibition of p38 MAPK with SB202190 (∼15%). Berberine treatment induced an increase in association between PKCα and PKA with KCNQ1 and produced phosphorylation of the channel. We conclude that berberine exerts its inhibitory effect on colonic Cl(-) secretion through inhibition of basolateral KCNQ1 channels responsible for K(+) recycling via a PKCα-dependent pathway.

6.
Methods Mol Biol ; 741: 471-88, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21594802

RESUMO

The cystic fibrosis transmembrane conductance regulator (CFTR) is the gene product mutated in cystic fibrosis, a common lethal genetic disease characterized by abnormal electrolyte transport across epithelia. CFTR functions as an ATP-gated, phosphorylation-regulated Cl- channel that mediates agonist-stimulated apical membrane epithelial Cl- and bicarbonate secretion and also regulates a variety of other transport proteins and cellular processes. CFTR belongs to the ATP-binding cassette (ABC) transporter superfamily. Its presumed architecture consists of two transmembrane domain regions that form the channel pore, two nucleotide-binding domains that bind and hydrolyze ATP, and a unique regulatory (R) domain that contains numerous protein kinase A (PKA) and protein kinase C (PKC) phosphorylation sites. Other kinases have also been shown more recently to phosphorylate and regulate CFTR activity. This chapter describes strategies and methods for studying the phosphorylation of CFTR both in vitro and whole-cell systems.


Assuntos
Bioquímica/métodos , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Extratos Celulares , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/química , Regulador de Condutância Transmembrana em Fibrose Cística/genética , DNA Complementar/genética , Humanos , Imunoprecipitação , Camundongos , Dados de Sequência Molecular , Fosforilação , Proteínas Quinases/metabolismo , Ratos , Transfecção
7.
Steroids ; 76(9): 867-76, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21600231

RESUMO

Excessive Cl(-) secretion is the driving force for secretory diarrhea. 17ß-Estradiol has been shown to inhibit Cl(-) secretion in rat distal colon through a nongenomic pathway. We examined whether 17ß-estradiol inhibits Cl(-) secretion in an animal model of secretory diarrhea and the downstream effectors involved. The effect of 17ß-estradiol on cholera toxin and heat-stable enterotoxin induced Cl(-) secretion in rat colonic mucosal sheets was studied by current-voltage clamping. Selective permeabilization of apical or basolateral membranes with amphotericin B or nystatin was used to isolate basolateral K(+) channel and apical Cl(-) channel activity, respectively. 17ß-Estradiol dose-dependently inhibited secretory responses to both toxins with IC(50) values of approximately 1nM. This effect was female-gender specific, with no inhibition observed in male tissues. 17ß-Estradiol responses were insensitive to the pure anti-estrogen ICI 182,720. 17ß-Estradiol exerted its effects downstream of enterotoxin-induced production of second messengers (cAMP and cGMP) but was dependent on PKCδ activation. In nystatin-permeabilized tissues, apical Cl(-) currents were unaffected by 17ß-estradiol treatment while basolateral K(+) current was profoundly inhibited by the hormone. This current was sensitive to the specific KCNQ1 channel inhibitors chromanol 293B and HMR-1556. In conclusion, 17ß-estradiol inhibits enterotoxin-induced Cl(-) secretion via a PKCδ-dependent mechanism involving inhibition of basolateral KCNQ1 channels. These data elucidate mechanisms of 17ß-estradiol inhibition of Cl(-) secretion induced by enterotoxins in intestinal epithelia, which may be relevant for the treatment of diarrheal diseases.


Assuntos
Toxinas Bacterianas/farmacologia , Cloretos/metabolismo , Toxina da Cólera/farmacologia , Colo/metabolismo , Enterotoxinas/farmacologia , Escherichia coli , Estradiol/farmacologia , Estrogênios/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Androgênios/farmacologia , Androgênios/fisiologia , Animais , Canais de Cloreto/metabolismo , Colo/citologia , Colo/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Ativação Enzimática , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Proteínas de Escherichia coli , Estradiol/fisiologia , Estrogênios/fisiologia , Feminino , Canal de Potássio KCNQ1/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Progesterona/farmacologia , Progesterona/fisiologia , Progestinas/farmacologia , Progestinas/fisiologia , Proteína Quinase C-delta/metabolismo , Ratos , Ratos Sprague-Dawley , Testosterona/farmacologia , Testosterona/fisiologia
8.
Am J Physiol Renal Physiol ; 299(6): F1308-19, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20861072

RESUMO

The KCNQ1 K(+) channel plays a key role in the regulation of several physiological functions, including cardiac excitability, cardiovascular tone, and body electrolyte homeostasis. The metabolic sensor AMP-activated protein kinase (AMPK) has been shown to regulate a growing number of ion transport proteins. To determine whether AMPK regulates KCNQ1, we studied the effects of AMPK activation on KCNQ1 currents in Xenopus laevis oocytes and collecting duct epithelial cells. AMPK activation decreased KCNQ1 currents and channel surface expression in X. laevis oocytes, but AMPK did not phosphorylate KCNQ1 in vitro, suggesting an indirect regulatory mechanism. As it has been recently shown that the ubiquitin-protein ligase Nedd4-2 inhibits KCNQ1 plasma membrane expression and that AMPK regulates epithelial Na(+) channels via Nedd4-2, we examined the role of Nedd4-2 in the AMPK-dependent regulation of KCNQ1. Channel inhibition by AMPK was blocked in oocytes coexpressing either a dominant-negative or constitutively active Nedd4-2 mutant, or a Nedd4-2 interaction-deficient KCNQ1 mutant, suggesting that Nedd4-2 participates in the regulation of KCNQ1 by AMPK. KCNQ1 is expressed at the basolateral membrane in mouse polarized kidney cortical collecting duct (mpkCCD(c14)) cells and in rat kidney. Treatment with the AMPK activators AICAR (2 mM) or metformin (1 mM) reduced basolateral KCNQ1 currents in apically permeabilized polarized mpkCCD(c14) cells. Moreover, AICAR treatment of rat kidney slices ex vivo induced AMPK activation and intracellular redistribution of KCNQ1 from the basolateral membrane in collecting duct principal cells. AICAR treatment also induced increased ubiquitination of KCNQ1 immunoprecipitated from kidney slice homogenates. These results indicate that AMPK inhibits KCNQ1 activity by promoting Nedd4-2-dependent channel ubiquitination and retrieval from the plasma membrane.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Animais , Células Epiteliais/efeitos dos fármacos , Células HEK293 , Humanos , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/metabolismo , Camundongos , Ubiquitina-Proteína Ligases Nedd4 , Ratos , Ribonucleotídeos , Proteínas de Xenopus , Xenopus laevis
9.
J Biol Chem ; 285(32): 24676-85, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20525692

RESUMO

The vacuolar H(+)-ATPase (V-ATPase) is a major contributor to luminal acidification in epithelia of Wolffian duct origin. In both kidney-intercalated cells and epididymal clear cells, cAMP induces V-ATPase apical membrane accumulation, which is linked to proton secretion. We have shown previously that the A subunit in the cytoplasmic V(1) sector of the V-ATPase is phosphorylated by protein kinase A (PKA). Here we have identified by mass spectrometry and mutagenesis that Ser-175 is the major PKA phosphorylation site in the A subunit. Overexpression in HEK-293T cells of either a wild-type (WT) or phosphomimic Ser-175 to Asp (S175D) A subunit mutant caused increased acidification of HCO(3)(-)-containing culture medium compared with cells expressing vector alone or a PKA phosphorylation-deficient Ser-175 to Ala (S175A) mutant. Moreover, localization of the S175A A subunit mutant expressed in HEK-293T cells was more diffusely cytosolic than that of WT or S175D A subunit. Acute V-ATPase-mediated, bafilomycin-sensitive H(+) secretion was up-regulated by a specific PKA activator in HEK-293T cells expressing WT A subunit in HCO(3)(-)-free buffer. In cells expressing the S175D mutant, V-ATPase activity at the membrane was constitutively up-regulated and unresponsive to PKA activators, whereas cells expressing the S175A mutant had decreased V-ATPase activity that was unresponsive to PKA activation. Finally, Ser-175 was necessary for PKA-stimulated apical accumulation of the V-ATPase in a polarized rabbit cell line of collecting duct A-type intercalated cell characteristics (Clone C). In summary, these results indicate a novel mechanism for the regulation of V-ATPase localization and activity in kidney cells via direct PKA-dependent phosphorylation of the A subunit at Ser-175.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Rim/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Sequência de Aminoácidos , Animais , Análise Mutacional de DNA , Humanos , Rim/fisiologia , Espectrometria de Massas/métodos , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Peptídeos/química , Fosforilação
10.
Am J Physiol Renal Physiol ; 299(1): F167-77, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20462973

RESUMO

The metabolic sensor AMP-activated protein kinase (AMPK) regulates several transport proteins, potentially coupling transport activity to cellular stress and energy levels. The creatine transporter (CRT; SLC6A8) mediates creatine uptake into several cell types, including kidney epithelial cells, where it has been proposed that CRT is important for reclamation of filtered creatine, a process critical for total body creatine homeostasis. Creatine and phosphocreatine provide an intracellular, high-energy phosphate-buffering system essential for maintaining ATP supply in tissues with high energy demands. To test our hypothesis that CRT is regulated by AMPK in the kidney, we examined CRT and AMPK distribution in the kidney and the regulation of CRT by AMPK in cells. By immunofluorescence staining, we detected CRT at the apical pole in a polarized mouse S3 proximal tubule cell line and in native rat kidney proximal tubules, a distribution overlapping with AMPK. Two-electrode voltage-clamp (TEV) measurements of Na(+)-dependent creatine uptake into CRT-expressing Xenopus laevis oocytes demonstrated that AMPK inhibited CRT via a reduction in its Michaelis-Menten V(max) parameter. [(14)C]creatine uptake and apical surface biotinylation measurements in polarized S3 cells demonstrated parallel reductions in creatine influx and CRT apical membrane expression after AMPK activation with the AMP-mimetic compound 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside. In oocyte TEV experiments, rapamycin and the AMPK activator 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranosyl 5'-monophosphate (ZMP) inhibited CRT currents, but there was no additive inhibition of CRT by ZMP, suggesting that AMPK may inhibit CRT indirectly via the mammalian target of rapamycin pathway. We conclude that AMPK inhibits apical membrane CRT expression in kidney proximal tubule cells, which could be important in reducing cellular energy expenditure and unnecessary creatine reabsorption under conditions of local and whole body metabolic stress.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Creatina/metabolismo , Células Epiteliais/enzimologia , Túbulos Renais Proximais/enzimologia , Proteínas de Membrana Transportadoras/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas da Membrana Plasmática de Transporte de Neurotransmissores/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Biotinilação , Western Blotting , Linhagem Celular Transformada , Polaridade Celular , Metabolismo Energético , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Células Epiteliais/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Cinética , Masculino , Potenciais da Membrana , Proteínas de Membrana Transportadoras/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Oócitos , Técnicas de Patch-Clamp , Proteínas da Membrana Plasmática de Transporte de Neurotransmissores/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ribonucleotídeos/farmacologia , Sirolimo/farmacologia , Sódio/metabolismo , Serina-Treonina Quinases TOR , Xenopus laevis
11.
Am J Physiol Renal Physiol ; 298(5): F1162-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20147366

RESUMO

The vacuolar H(+)-ATPase (V-ATPase) in type A kidney intercalated cells is a major contributor to acid excretion in the collecting duct. The mechanisms of V-ATPase-trafficking regulation in kidney intercalated cells have not been well-characterized. In developmentally related epididymal clear cells, we showed previously that PKA, acting downstream of soluble adenylyl cyclase (sAC), induces V-ATPase apical membrane accumulation. These PKA-mediated effects were inhibited by activators of the metabolic sensor AMP-activated kinase (AMPK) in clear cells. Here, we examined the regulation of V-ATPase subcellular localization in intercalated cells by PKA and AMPK in rat kidney tissue slices ex vivo. Immunofluorescence labeling of kidney slices revealed that the PKA activator N(6)-monobutyryl cAMP (6-MB-cAMP) induced V-ATPase apical membrane accumulation in collecting duct intercalated cells, whereas the V-ATPase had a more cytosolic distribution when incubated in Ringer buffer alone for 30 min. V-ATPase accumulated at the apical membrane in intercalated cells in kidney slices incubated in Ringer buffer for 75 min, an effect that was prevented by treatment with PKA inhibitor (mPKI). The V-ATPase distribution was cytosolic in intercalated cells treated with the carbonic anhydrase inhibitor acetazolamide or the sAC inhibitor KH7, effects that were overridden by 6-MB-cAMP. Preincubation of kidney slices with an AMPK activator blocked V-ATPase apical membrane accumulation induced by 6-MB-cAMP, suggesting that AMPK antagonizes cAMP/PKA effects on V-ATPase distribution. Taken together, our results suggest that in intercalated cells V-ATPase subcellular localization and therefore its activity may be coupled to acid-base status via PKA, and metabolic status via AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Rim/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Anidrases Carbônicas/metabolismo , AMP Cíclico/metabolismo , Rim/citologia , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley
12.
Mol Endocrinol ; 23(11): 1885-99, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19846538

RESUMO

The secretion of Cl(-) across distal colonic crypt cells provides the driving force for the movement of fluid into the luminal space. 17beta-Estradiol (E2) produces a rapid and sustained reduction in secretion in females, which is dependent on the novel protein kinase C delta (PKC delta) isozyme and PKA isoform I targeting of KCNQ1 channels. This sexual dimorphism in the E2 response is associated with a higher expression level of PKC delta in female compared with the male tissue. The present study revealed the antisecretory response is regulated throughout the female reproductive (estrous) cycle and is primed by genomic regulation of the kinases. E2 (1-10 nm) decreased cAMP-dependent secretion in colonic epithelia during the estrus, metestrus, and diestrus stages. A weak inhibition of secretion was demonstrated in the proestrus stage. The expression levels of PKC delta and PKA fluctuated throughout the estrous cycle and correlated with the potency of the antisecretory effect of E2. The expression of PKC delta and PKA were up-regulated by estrogen at a transcriptional level via a PKC delta-MAPK-cAMP response element-binding protein-regulated pathway indicating a genomic priming of the antisecretory response. PK Cdelta was activated by the membrane-impermeant E2-BSA, and this response was inhibited by the estrogen receptor antagonist ICI 182,780. The 66-kDa estrogen receptor-alpha isoform was present at the plasma membrane of female colonic crypt cells with a lower abundance found in male colonic crypts. The study demonstrates estrogen regulation of intestinal secretion both at a rapid and transcriptional level, demonstrating an interdependent relationship between both nongenomic and genomic hormone responses.


Assuntos
Colo/metabolismo , Estrogênios/metabolismo , Ciclo Estral/metabolismo , Genômica , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estradiol/metabolismo , Feminino , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Fosforilação , Isoformas de Proteínas , Proteína Quinase C-delta/metabolismo , RNA Mensageiro/metabolismo , Ratos , Transcrição Gênica
13.
Am J Physiol Cell Physiol ; 296(4): C672-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19211918

RESUMO

Acidic luminal pH and low [HCO(3)(-)] maintain sperm quiescent during maturation in the epididymis. The vacuolar H(+)-ATPase (V-ATPase) in clear cells is a major contributor to epididymal luminal acidification. We have shown previously that protein kinase A (PKA), acting downstream of soluble adenylyl cyclase stimulation by alkaline luminal pH or HCO(3)(-), induces V-ATPase apical membrane accumulation in clear cells. Here we examined whether the metabolic sensor AMP-activated protein kinase (AMPK) regulates this PKA-induced V-ATPase apical membrane accumulation. Immunofluorescence labeling of rat and non-human primate epididymides revealed specific AMPK expression in epithelial cells. Immunofluorescence labeling of rat epididymis showed that perfusion in vivo with the AMPK activators 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) or A-769662 induced a redistribution of the V-ATPase into subapical vesicles, even in the presence of a luminal alkaline (pH 7.8) buffer compared with that of controls perfused without drug. Moreover, preperfusion with AICAR blocked the PKA-mediated V-ATPase translocation to clear cell apical membranes induced by N(6)-monobutyryl-cAMP (6-MB-cAMP). Purified PKA and AMPK both phosphorylated V-ATPase A subunit in vitro. In HEK-293 cells [(32)P]orthophosphate in vivo labeling of the A subunit increased following PKA stimulation and decreased following RNA interference-mediated knockdown of AMPK. Finally, the extent of PKA-dependent in vivo phosphorylation of the A subunit increased with AMPK knockdown. In summary, our findings suggest that AMPK inhibits PKA-mediated V-ATPase apical accumulation in epididymal clear cells, that both kinases directly phosphorylate the V-ATPase A subunit in vitro and in vivo, and that AMPK inhibits PKA-dependent phosphorylation of this subunit. V-ATPase activity may be coupled to the sensing of acid-base status via PKA and to metabolic status via AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Epididimo/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Compostos de Bifenilo , Bucladesina/análogos & derivados , Bucladesina/metabolismo , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativadores de Enzimas/farmacologia , Epididimo/citologia , Epididimo/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Macaca mulatta , Masculino , Camundongos , Fosforilação , Transporte Proteico , Pironas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Ribonucleotídeos/farmacologia , Tiofenos/farmacologia , Transfecção , ATPases Vacuolares Próton-Translocadoras/genética
14.
Steroids ; 73(9-10): 885-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18329060

RESUMO

The non-genomic action of steroid hormones regulates a wide variety of cellular responses including regulation of ion transport, cell proliferation, migration, death and differentiation. In order to achieve such plethora of effects steroid hormones utilize nearly all known signal transduction pathways. One of the key signalling molecules regulating the non-genomic action of steroid hormones is protein kinase C (PKC). It is thought that rapid action of steroids hormones results from the activation of plasma membrane receptors; however, their molecular identity remains elusive. In recent years, an increasing number of studies have pointed at the selective binding and activation of specific PKC isoforms by steroid hormones. This has led to the hypothesis that PKC could act as a receptor as well as a transducer of the non-genomic effects of these hormones. In this review we summarize the current knowledge of the direct binding and activation of PKC by steroid hormones.


Assuntos
Proteína Quinase C/química , Proteína Quinase C/metabolismo , Esteroides/metabolismo , Aldosterona/metabolismo , Animais , Sítios de Ligação , Estradiol/metabolismo , Humanos , Modelos Biológicos , Mutação , Ligação Proteica , Isoformas de Proteínas
15.
J Steroid Biochem Mol Biol ; 108(3-5): 310-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17951051

RESUMO

Aldosterone is a crucial modulator of ion transport across high resistance epithelia and regulates whole body electrolyte balance through its effects on the kidney and colon. The net consequence of aldosterone release is to promote salt conservation. The genomic mechanism of aldosterone action is relatively well characterized and the role of the classical mineralocorticoid receptor as a ligand-dependent transcription factor is well established. The rapid effects of aldosterone on target tissues are less well understood and there is still controversy over the identity of the aldosterone non-genomic receptor. Greater understanding of the physiological consequences of aldosterone's rapid responses in the kidney and colon has been achieved through the identification of definite and putative membrane targets and their signaling regulators.


Assuntos
Aldosterona/fisiologia , Colo/efeitos dos fármacos , Rim/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/fisiologia , Animais , Sinalização do Cálcio , Colo/fisiologia , Canais Epiteliais de Sódio/fisiologia , Humanos , Rim/fisiologia , Canais de Potássio/fisiologia , Proteínas Quinases/fisiologia , ATPases Translocadoras de Prótons/fisiologia , Receptores de Mineralocorticoides/fisiologia , ATPase Trocadora de Sódio-Potássio/fisiologia , Fatores de Tempo
16.
Mol Endocrinol ; 21(11): 2637-50, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17666590

RESUMO

Protein kinase C (PKC) is a signal transduction protein that has been proposed to mediate rapid responses to steroid hormones. Previously, we have shown aldosterone directly activates PKCalpha whereas 17beta-estradiol activates PKCalpha and PKCdelta; however, neither the binding to PKCs nor the mechanism of action has been established. To determine the domains of PKCalpha and PKCdelta involved in binding of aldosterone and 17beta-estradiol, glutathione S-transferase fusion recombinant PKCalpha and PKCdelta mutants were used to perform in vitro binding assays with [(3)H]aldosterone and [(3)H]17beta-estradiol. 17beta-Estradiol bound both PKCalpha and PKCdelta but failed to bind PKC mutants lacking a C2 domain. Similarly, aldosterone bound only PKCalpha and mutants containing C2 domains. Thus, the C2 domain is critical for binding of these hormones. Binding affinities for aldosterone and 17beta-estradiol were between 0.5-1.0 nM. Aldosterone and 17beta-estradiol competed for binding to PKCalpha, suggesting they share the same binding site. Phorbol 12,13-dybutyrate did not compete with hormone binding; furthermore, they have an additive effect on PKC activity. EC(50) for activation of PKCalpha and PKCdelta by aldosterone and 17beta-estradiol was approximately 0.5 nM. Immunoblot analysis using a phospho-PKC antibody revealed that upon binding, PKCalpha and PKCdelta undergo autophosphorylation with an EC(50) in the 0.5-1.0 nm range. 17beta-Estradiol activated PKCalpha and PKCdelta in estrogen receptor-positive and -negative breast cancer cells (MCF-7 and HCC-38, respectively), suggesting estrogen receptor expression is not required for 17beta-estradiol-induced PKC activation. The present results provide first evidence for direct binding and activation of PKCalpha and PKCdelta by steroid hormones and the molecular mechanisms involved.


Assuntos
Aldosterona/química , Estradiol/metabolismo , Proteína Quinase C/química , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática , Ésteres/química , Glutationa Transferase/metabolismo , Humanos , Cinética , Mutação , Fosforilação , Ligação Proteica , Isoformas de Proteínas , Proteínas Recombinantes de Fusão/química , Esteroides/química , Esteroides/metabolismo
17.
J Biol Chem ; 282(34): 24563-73, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17556370

RESUMO

The estrogen sex steroid 17beta-estradiol rapidly inhibits secretagogue-stimulated cAMP-dependent Cl(-) secretion in the female rat distal colonic crypt by the inhibition of basolateral K(+) channels. In Ussing chamber studies, both the anti-secretory response and inhibition of basolateral K(+) current was shown to be attenuated by pretreatment with rottlerin, a PKCdelta-specific inhibitor. In whole cell patch-clamp analysis, 17beta-estradiol inhibited a chromanol 293B-sensitive KCNQ1 channel current in isolated female rat distal colonic crypts. Estrogen had no effect on KCNQ1 channel currents in colonic crypts isolated from male rats. Female distal colonic crypts expressed a significantly higher amount of PKCdelta in comparison to male tissue. PKCdelta and PKA were activated at 5 min in response to 17beta-estradiol in female distal colonic crypts only. Both PKCdelta- and PKA-associated with the KCNQ1 channel in response to 17beta-estradiol in female distal colonic crypts, and no associations were observed in crypts from males. PKA activation, association with KCNQ1, and phosphorylation of the channel were regulated by PKCdelta as the responses were blocked by pretreatment with rottlerin. Taken together, our experiments have identified the molecular targets underlying the anti-secretory response to estrogen involving the inhibition of KCNQ1 channel activity via PKCdelta- and PKA-dependent signaling pathways. This is a novel gender-specific mechanism of regulation of an ion channel by estrogen. The anti-secretory response described in this study provides molecular insights whereby estrogen causes fluid retention effects in the female during periods of high circulating plasma estrogen levels.


Assuntos
Colo/metabolismo , Estradiol/metabolismo , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/fisiologia , Animais , Transporte Biológico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estrogênios/sangue , Estrogênios/metabolismo , Feminino , Masculino , Modelos Biológicos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais
18.
Endocrinology ; 144(4): 1266-72, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12639909

RESUMO

Aldosterone increases Na(+),K(+)-adenosine triphophatase (Na(+),K(+)-ATPase) pump activity and abundance under chronic conditions in several tissues, including rat arterial vessels. The present study was undertaken to evaluate whether aldosterone has also short-term effects on the Na(+),K(+)-ATPase of rat aorta. The pump function was measured as ouabain-sensitive (86)Rb/K uptake in aortic rings. Addition of aldosterone induced a rapid inhibition of the Na(+),K(+)-ATPase (57.0 +/- 2.3% of control values; P < 0.05; n = 8), followed by a return to control values after 120 min. The aldosterone-induced decrease in ouabain-sensitive (86)Rb/K uptake was prevented by the new mineralocorticoid receptor antagonist eplerenone. The inhibition of gene transcription (actinomycin D) or protein synthesis (cycloheximide) had no effect on short-term aldosterone action on Na(+),K(+)-ATPase. The rapid aldosterone inhibition was also observed in the presence of monensin, a sodium-specific ionophore. Rapamycin, an immunosuppressive drug that stabilizes the heat shock protein-steroid receptor complex, blocked the rapid aldosterone effect. Bisindole I, an inhibitor of protein kinase C, also blocked nongenomic action of aldosterone on the Na pump. The nongenomic effect of aldosterone was inhibited by disrupters of microtubule (colchicine). Plasma membrane protein biotinylation of aortic segments and Western blot indicated a diminished presence of catalytic isoforms of Na(+),K(+)-ATPase on the cell surface. Our findings indicate that aldosterone has a nongenomic effect on the Na(+),K(+)-ATPase of vascular tissue. This effect is mediated through protein kinase C activation and implies reduced cell surface abundance of catalytic subunits. These observations together with our previous report on chronic hormone replacement suggest that aldosterone is directly involved in ionic cellular homeostasis of the vascular system through Na pump regulation.


Assuntos
Aldosterona/farmacologia , Aorta Torácica/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Espironolactona/análogos & derivados , Animais , Domínio Catalítico , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Inibidores Enzimáticos/farmacologia , Eplerenona , Indóis/farmacologia , Ionóforos/farmacologia , Masculino , Maleimidas/farmacologia , Antagonistas de Receptores de Mineralocorticoides , Monensin/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/química , Espironolactona/farmacologia , Transcrição Gênica/efeitos dos fármacos
19.
Biochim Biophys Acta ; 1566(1-2): 116-28, 2002 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-12421543

RESUMO

Fifty years ago, Hans Ussing described the mechanism by which ions are actively transported across frog skin. Since then, an enormous amount of effort has been invested in determining the cellular and molecular specifics of the transport mechanisms and their regulatory pathways. Ion transport in high-resistance epithelia is regulated by a variety of hormonal and non-hormonal factors. In vertebrates, steroid hormones such as mineralocorticoids, glucocorticoids and estrogens are major regulators of ion and water transport and hence are central to the control of extracellular fluid volume and blood pressure. Steroid hormones act through nuclear receptors to control the transcriptional activity of specific target genes, such as ion channels, ion transporters and ion pumps. These effects are observed after a latency of several hours and can last for days leading to cellular differentiation that allows a higher transport activity. This pathway is the so-called genomic phase. However, in the past 10 years, it has become apparent that steroid hormones can regulate electrolyte and water transport in tight epithelia independently of the transcription of these ion channels and transporters by regulating ion transporter activity in a non-genomic fashion via modulation of various signal transduction pathways. The molecular mechanisms underlying the steroid hormone-induced activation of signal transduction pathways such as protein kinase C (PKC), protein kinase A (PKA), intracellular calcium, intracellular pH and mitogen-activated protein kinases (MAPKs) and how non-genomic activation of these pathways influences epithelial ion transport will be discussed in this review.


Assuntos
Colo/efeitos dos fármacos , Hormônios/farmacologia , Pele/efeitos dos fármacos , Animais , Anuros , Cloretos/metabolismo , Colo/metabolismo , Ativação Enzimática/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Estrogênios/farmacologia , Glucocorticoides/farmacologia , Humanos , Transporte de Íons/efeitos dos fármacos , Mineralocorticoides/farmacologia , Proteína Quinase C/metabolismo , Fatores Sexuais , Transdução de Sinais/efeitos dos fármacos , Pele/metabolismo , Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/efeitos dos fármacos
20.
Steroids ; 67(6): 483-91, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11960625

RESUMO

Studies from our laboratory have demonstrated rapid ( < 1 min) non-genomic activation of Na(+)-H(+) exchange, K(+) recycling, PKC activity and a PKC-dependent Ca(2+) entry through L-type Ca(2+) channels specifically by mineralocorticoids in distal colon. Aldosterone directly stimulates the activity of the PKC alpha isoform (but not PKC delta, PKC epsilon and PKC zeta) in a cell-free assay system containing only purified commercially available enzyme, appropriate substrate peptide, co-factors and lipid vesicles. The primary ion transport target of the non-genomic signal transduction cascade elicited by aldosterone in epithelia is the Na(+)-H(+) exchanger. In isolated colonic crypts, aldosterone produced a PKC alpha sensitive intracellular alkalinisation within 1 min of hormone addition. Intracellular alkalinisation upregulates an ATP-dependent K(+) channel, which is involved in K(+) recycling to maintain the electrical driving force for Na(+) absorption, while inhibiting a Ca(2+) -dependent K(+) channel, which generates the charge balance for Cl(-) secretion. The non-genomic response to aldosterone in distal colon appears to enhance the capacity for absorption while down-regulating the potential for secretion. We have also demonstrated rapid (< 1 min) non-genomic activation of Na(+)-H(+) exchange, K(+) recycling, PKC alpha activity, and a PKC delta- and PKA-dependent Ca(2+) entry through di-hydropyridine-blockable Ca(2+) channels specifically by 17beta-estradiol in distal colon. These rapid effects are female gender specific and are insensitive to inhibitors of the classical estrogen receptor (ER). 17 beta-Estradiol directly stimulated the activity of both PKC delta and PKC alpha (but not PKC epsilon or PKC zeta) in a cell-free assay system. E2 rapidly inhibited basolateral K(Ca) channel activity which would be expected to result in an acute inhibition of Cl(-) secretion. Physiological concentrations of E2 (0.1-10 nM) reduced both basal and secretagogue-induced Cl(-) secretion. This anti-secretory effect of E2 is sensitive to PKC inhibition, intracellular Ca(2+) chelation, and is female gender specific and insensitive to inhibitors of the classical ER. These observations link rapid non-genomic activation of second messengers with a rapid gender-specific physiological effect in the whole tissue. Aldosterone and E2 differ in their protein kinase signal transduction and both hormones stimulate specific PKC isoforms indicating both common and divergent signalling systems for salt-retaining steroid hormones. The physiological function of non-genomic effects of aldosterone and estradiol is to shift the balance from net secretion to net absorption in a pluripotential epithelium.


Assuntos
Aldosterona/farmacologia , Canais de Cálcio/metabolismo , Estradiol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/efeitos dos fármacos , Animais , Canais de Cálcio/efeitos dos fármacos , Colo/metabolismo , Ativação Enzimática/efeitos dos fármacos , Epitélio/metabolismo , Feminino , Humanos , Isoenzimas/metabolismo , Masculino , Mamíferos , Fosfolipases A/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Ratos , Transdução de Sinais/fisiologia , Trocadores de Sódio-Hidrogênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA