Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
J Biol Chem ; 291(47): 24787-24799, 2016 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-27733684

RESUMO

Hypotonic stress decreased claudin-1 and -2 expression levels in renal tubular epithelial HK-2 and Madin-Darby canine kidney cells. Here, we examined the regulatory mechanism involved in this decrease. The hypotonicity-induced decrease in claudin expression was inhibited by the following: SB202190, a p38 MAPK inhibitor, but not by U0126, a MEK inhibitor; Go6983, a protein kinase C inhibitor; or SP600125, a Jun N-terminal protein kinase inhibitor. Hypotonic stress increased transepithelial electrical resistance, which was inhibited by SB202190. The mRNA expression level of claudin-1 was decreased by hypotonic stress but that of claudin-2 was not. Hypotonic stress decreased the protein stability of claudin-1 and -2. The hypotonicity-induced decrease in claudin expression was inhibited by the following: chloroquine, a lysosome inhibitor; dynasore and monodansylcadaverine, clathrin-dependent endocytosis inhibitors; and siRNA against clathrin heavy chain. Claudin-1 and -2 were mainly distributed in the cytosol and tight junctions (TJs) in the chloroquine- and monodansylcadaverine-treated cells, respectively. Hypotonic stress decreased the phosphorylation levels of claudin-1 and -2, which were inhibited by the protein phosphatase inhibitors okadaic acid and cantharidin. Dephosphorylated mutants of claudin-1 and -2 were mainly distributed in the cytosol, which disappeared in response to hypotonic stress. In contrast, mimicking phosphorylation mutants were distributed in the TJs, which were not decreased by hypotonic stress. We suggest that hypotonic stress induces dephosphorylation, clathrin-dependent endocytosis, and degradation of claudin-1 and -2 in lysosomes, resulting in disruption of the TJ barrier in renal tubular epithelial cells.


Assuntos
Clatrina/metabolismo , Claudina-1/metabolismo , Claudinas/metabolismo , Regulação para Baixo , Endocitose , Células Epiteliais/metabolismo , Túbulos Renais Distais/metabolismo , Pressão Osmótica , Animais , Clatrina/genética , Claudina-1/genética , Claudinas/genética , Citosol/metabolismo , Cães , Humanos , Túbulos Renais Distais/citologia , Células Madin Darby de Rim Canino , Fosforilação , Junções Íntimas/genética , Junções Íntimas/metabolismo
2.
Biochim Biophys Acta ; 1863(6 Pt A): 1170-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26919807

RESUMO

Abnormal expression of claudin subtypes has been reported in various cancers. However, the pathological role of each claudin has not been clarified in detail. Claudin-18 was absent in human non-small cell and small cell lung cancers, although it is expressed in normal lung tissues. Here, we examined the effect of claudin-18 expression on the expression of junctional proteins, cell proliferation, and cell motility using human lung adenocarcinoma A549 cells. Real-time PCR and western blotting showed that exogenous expression of claudin-18 had no effect on the expression of junctional proteins including claudin-1, zonula occludens-1 (ZO-1), occludin, and E-cadherin. Claudin-18 was mainly distributed in cell-cell contact areas concomitant with ZO-1. Cell proliferation was significantly decreased at 48 and 72h after seeding of claudin 18-expressing cells. Claudin-18 suppressed cell motility, whereas it increased cell death in anoikis. Claudin-18 decreased phosphorylated (p)-3-phosphoinositide-dependent protein kinase-1 (PDK1) and p-Akt levels without affecting p-epidermal growth factor receptor and p-phosphatidylinositol-3 kinase (PI3K) levels. Furthermore, claudin-18 was bound with PDK1 and suppressed the nuclear localization of PDK1. We suggest that claudin-18 suppresses the abnormal proliferation and motility of lung epithelial cells mediated by inhibition of the PI3K/PDK1/Akt signaling pathway.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Claudinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Anoikis/efeitos dos fármacos , Anoikis/fisiologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cromonas/farmacologia , Claudinas/genética , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Microscopia Confocal , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Piruvato Desidrogenase Quinase de Transferência de Acetil , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/metabolismo
3.
Biochim Biophys Acta ; 1848(10 Pt A): 2326-36, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26163137

RESUMO

Claudins are tight junctional proteins and comprise a family of over 20 members. Abnormal expression of claudins is reported to be involved in tumor progression. Claudin-2 is highly expressed in lung adenocarcinoma tissues and increases cell proliferation, whereas it is not expressed in normal tissues. Claudin-2-targeting molecules such as peptides and small molecules may be novel anti-cancer drugs. The short peptide with the sequence DFYSP, which mimics the second extracellular loop of claudin-2, decreased claudin-2 content in the cytoplasmic fraction of A549 cells. In contrast, it did not affect the content in the nuclear fraction. The decrease in claudin-2 content was inhibited by chloroquine (CQ), a lysosomal inhibitor, but not by MG-132, a proteasome inhibitor. In the presence of DFYSP peptide and CQ, claudin-2 was co-localized with LAMP-1, a lysosomal marker. The DFYSP peptide-induced decrease in claudin-2 content was inhibited by monodancylcadaverine (MDC), an inhibitor of clathrin-dependent endocytosis. DFYSP peptide increased lysosome content and cathepsin B release, and induced cellular injury, which were inhibited by MDC. Cellular injury induced by DFYSP peptide was inhibited by necrostatin-1, an inhibitor of necrotic cell death, but not by Z-VAD-FMK, an inhibitor of apoptotic cell death. Our data indicate that DFYSP peptide increases the accumulation of the peptide and claudin-2 into the lysosome, resulting in lysosomal damage. Claudin-2 may be a new target for lung cancer therapy.


Assuntos
Clatrina/metabolismo , Claudina-2/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Lisossomos/metabolismo , Peptídeos/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Endocitose/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/patologia
4.
Hepatology ; 62(4): 1215-26, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25999152

RESUMO

UNLABELLED: The adenosine triphosphate-binding cassette (ABC) half-transporters Abcg5 and Abcg8 promote the secretion of neutral sterol into bile. Studies have demonstrated the diet-induced gene expression of these transporters, but the regulation of their trafficking when the nutritional status changes in the liver remains to be elucidated. Here, we generated a novel in vivo kinetic analysis that can monitor the intracellular trafficking of Abcg5/Abcg8 in living mouse liver by in vivo transfection of the genes of fluorescent protein-tagged transporters and investigated how hypernutrition affects the canalicular trafficking of these transporters. The kinetic analysis showed that lithogenic diet consumption accelerated the translocation of newly synthesized fluorescent-tagged transporters to intracellular pools in an endosomal compartment and enhanced the recruitment of these pooled gene products into the bile canalicular membrane in mouse liver. Because some ABC transporters are reported to be recruited from intracellular pools to the bile canaliculi by cyclic adenosine monophosphate (cAMP) signaling, we next evaluated the involvement of this machinery in a diet-induced event. Administration of a protein kinase A inhibitor, N-(2-{[3-(4-bromophenyl)-2-propenyl]amino}ethyl)-5-isoquinolinesulfonamide, decreased the canalicular expression of native Abcg5/Abcg8 in lithogenic diet-fed mice, and injection of a cAMP analog, dibutyryl cAMP, transiently increased their levels in standard diet-fed mice, indicating the involvement of cAMP signaling. Indeed, canalicular trafficking of the fluorescent-tagged Abcg5/Abcg8 was enhanced by dibutyryl cAMP administration. CONCLUSION: These observations suggest that diet-induced lipid loading into liver accelerates the trafficking of Abcg5/Abcg8 to the bile canalicular membrane through cAMP signaling machinery.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Canalículos Biliares/fisiologia , AMP Cíclico/fisiologia , Lipoproteínas/fisiologia , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Dieta , Cinética , Camundongos , Transporte Proteico , Transdução de Sinais
5.
Drug Metab Dispos ; 44(1): 137-50, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26534988

RESUMO

The human pregnane X receptor (hPXR) is a xenobiotic-sensing nuclear receptor that transcriptionally regulates drug metabolism-related genes. The aim of the present study was to elucidate the mechanism by which hPXR is regulated through threonine-408. A phosphomimetic mutation at threonine-408 (T408D) reduced the transcriptional activity of hPXR and its protein stability in HepG2 and SW480 cells in vitro and mouse livers in vivo. Proteasome inhibitors (calpain inhibitor I and MG132) and Hsp90 inhibitor geldanamycin, but not Hsp70 inhibitor pifithrin-µ, increased wild-type (WT) hPXR in the nucleus. The translocation of the T408D mutant to the nucleus was significantly reduced even in the presence of proteasome inhibitors, whereas the complex of yellow fluorescent protein (YFP)-hPXR T408D mutant with heat shock cognate protein 70/heat shock protein 70 and carboxy terminus Hsp70-interacting protein (CHIP; E3 ligase) was similar to that of the WT in the cytoplasm. Treatment with pifithrin-µ and transfection with anti-CHIP small-interfering RNA reduced the levels of CYP3A4 mRNA induced by rifampicin, suggesting the contribution of Hsp70 and CHIP to the transactivation of hPXR. Autophagy inhibitor 3-methyladenine accumulated YFP-hPXR T408D mutant more efficiently than the WT in the presence of proteasome inhibitor lactacystin, and the T408D mutant colocalized with the autophagy markers, microtubule-associated protein 1 light chain 3 and p62, which were contained in the autophagic cargo. Lysosomal inhibitor chloroquine caused the marked accumulation of the T408D mutant in the cytoplasm. Protein kinase C (PKC) directly phosphorylated the threonine-408 of hPXR. These results suggest that hPXR is regulated through its phosphorylation at threonine-408 by PKC, CHIP/chaperone-dependent stability check, and autophagic degradation pathway.


Assuntos
Autofagia , Hepatócitos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Esteroides/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Citocromo P-450 CYP3A/biossíntese , Citocromo P-450 CYP3A/genética , Indução Enzimática , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos ICR , Mutação , Fosforilação , Receptor de Pregnano X , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma/farmacologia , Proteína Quinase C/metabolismo , Estabilidade Proteica , Proteólise , Interferência de RNA , Receptores de Esteroides/química , Receptores de Esteroides/genética , Treonina , Fatores de Tempo , Transcrição Gênica , Transfecção , Ubiquitina-Proteína Ligases/genética
6.
J Biol Chem ; 289(19): 13112-23, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24659781

RESUMO

Claudin-16 (CLDN16) regulates the paracellular reabsorption of Mg(2+) in the thick ascending limb of Henle's loop. However, the mechanism regulating the tight junctional localization of CLDN16 remains unknown. In yeast two-hybrid systems, we found that CLDN16 bound to syntaxin 8 (STX8), a target soluble N-ethylmaleimide-sensitive factor attachment protein receptor. We have examined the effect of STX8 on the localization and function of CLDN16 using Madin-Darby canine kidney cells expressing FLAG-tagged CLDN16. A pulldown assay showed that the carboxyl cytoplasmic region of human CLDN16 bound to STX8. CLDN16 was localized in the thick ascending limb, whereas STX8 was widely distributed throughout the rat kidney. An association between CLDN16 and STX8 was observed in rat renal homogenates and Madin-Darby canine kidney cells. STX8 siRNA decreased the cell surface localization of CLDN16 and transepithelial electrical resistance and permeability to Mg(2+) but increased the co-localization of CLDN16 with early endosome and lysosome markers. Dephosphorylation of CLDN16 by protein kinase A inhibitors and S217A mutant, a dephosphorylated form, decreased the association with STX8 and the cell surface localization of CLDN16. Recycling assays indicated that STX8 siRNA decreased the trafficking of CLDN16 to the plasma membrane without affecting endocytosis. Dominant negative Rab11 and recycling inhibitor primaquine decreased the cell surface localization of CLDN16, which was similar to that in STX8 siRNA-transfected cells. These results suggest that STX8 mediates the recycling of CLDN16 and constitutes an important component of the CLDN16 trafficking machinery in the kidney.


Assuntos
Claudinas/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Qa-SNARE/metabolismo , Junções Íntimas/metabolismo , Substituição de Aminoácidos , Animais , Claudinas/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Cães , Endossomos/genética , Endossomos/metabolismo , Células Epiteliais/citologia , Humanos , Túbulos Renais Proximais/citologia , Lisossomos/genética , Lisossomos/metabolismo , Células Madin Darby de Rim Canino , Masculino , Mutação de Sentido Incorreto , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia , Proteínas Qa-SNARE/genética , Ratos , Ratos Wistar , Junções Íntimas/genética
7.
Biochim Biophys Acta ; 1843(9): 2079-88, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24907662

RESUMO

Claudin-2 is expressed in human lung adenocarcinoma tissue and cell lines, although it is absent in normal lung tissue. However, the role of claudin-2 in cell proliferation and the regulatory mechanism of intracellular distribution remain undefined. Proliferation of human adenocarcinoma A549 cells was decreased by claudin-2 knockdown together with a decrease in the percentage of S phase cells. This knockdown decreased the expression levels of ZONAB and cell cycle regulators. Claudin-2 was distributed in the nucleus in human adenocarcinoma tissues and proliferating A549 cells. The nuclear distribution of ZONAB and percentage of S phase cells were higher in cells exogenously expressing claudin-2 with a nuclear localization signal than in cells expressing claudin-2 with a nuclear export signal. Nuclear claudin-2 formed a complex with ZO-1, ZONAB, and cyclin D1. Nuclear distribution of S208A mutant, a dephosphorylated form of claudin-2, was higher than that of wild type. We suggest that nuclear distribution of claudin-2 is up-regulated by dephosphorylation and claudin-2 serves to retain ZONAB and cyclin D1 in the nucleus, resulting in the enhancement of cell proliferation in lung adenocarcinoma cells.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Núcleo Celular/metabolismo , Claudinas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Adenocarcinoma de Pulmão , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colforsina/farmacologia , Ciclina D1/metabolismo , Fase G1/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Mutantes/metabolismo , Sinais de Exportação Nuclear , Sinais de Localização Nuclear/metabolismo , Fosforilação/efeitos dos fármacos , Fase S/efeitos dos fármacos , Fatores de Tempo , Proteína da Zônula de Oclusão-1/metabolismo
8.
J Cell Physiol ; 230(11): 2776-87, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25825272

RESUMO

Hyperosmolarity decreases claudin-2 expression in renal tubular epithelial cells, but the molecular mechanism remains undefined. Here, we found that the hyperosmolarity-induced decrease in claudin-2 expression is inhibited by Go6983, a non-selective protein kinase C (PKC) inhibitor, and PKCß specific inhibitor in Madin-Darby canine kidney II cells. Hyperosmolarity increased intracellular free Ca(2+) concentration and phosphorylated PKCß level, which were inhibited by RN-1734, an antagonist of transient receptor potential vanilloid 4 channel. Phorbol 12-myristate 13-acetate, a PKC activator, decreased claudin-2 expression. These results indicate hyperosmolarity decreases claudin-2 expression mediated by the activation of RN-1734-sensitive channel and PKCß. Hyperosmolarity decreased promoter activity of claudin-2, which was inhibited by Go6983 and PKCß inhibitor similar to those in real-time PCR and Western blotting. The effect of hyperosmolarity on promoter activity was not observed in the construct of -469/-6, a deletion mutant. Claudin-2 has hyperosmolarity-sensitive region in its promoter, which includes GATA binding site. Hyperosmolarity decreased the nuclear level of GATA-2, which was inhibited by Go6983 and PKCß inhibitor. Mutation of GATA binding site decreased the basal promoter activity and inhibited the effect of hyperosmolarity. In contrast, the hyperosmolarity-induced decrease in reporter activity and claudin-2 expression were rescued by over-expression of wild type GATA-2. Chromatin immunoprecipitation assay showed that GATA-2 bound to promoter region of claudin-2. These results suggest that hyperosmolarity decreases the expression level of claudin-2 via a decrease in PKCß-dependent GATA-2 transcriptional activity in renal tubular epithelial cells.


Assuntos
Claudina-2/biossíntese , Fator de Transcrição GATA2/biossíntese , Concentração Osmolar , Proteína Quinase C beta/biossíntese , Animais , Sítios de Ligação , Sinalização do Cálcio/efeitos dos fármacos , Claudina-2/genética , Cães , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Indóis/administração & dosagem , Túbulos Renais Proximais/metabolismo , Células Madin Darby de Rim Canino , Maleimidas/administração & dosagem , Regiões Promotoras Genéticas , Proteína Quinase C beta/antagonistas & inibidores , Ratos , Sulfonamidas , Acetato de Tetradecanoilforbol/administração & dosagem
9.
FASEB J ; 28(1): 440-52, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24100020

RESUMO

Platelet-activating factor receptor (PAFR)-deficient mice developed a more severe obese state characterized by higher body mass (~25%) and epididymal fat mass (~55%) with age than that of wild-type (WT) littermates. PAFR-deficient mice did not show changes in the expression of critical genes involved in anabolic and catabolic metabolism in adipose, liver, and muscle tissues between 6 and 36 wk. However, a 38-81% reduction in ß3/ß1-adrenergic receptor (AR) and uncoupling protein 1 (UCP1) mRNA and protein levels was observed in the interscapular brown adipose tissue (BAT) of PAFR-deficient mice. Whereas a single injection of the ß3-adrenergic agonist, CL-316,243 (25 µg/kg) increased temperatures in the brown fat and rectums of WT mice, this increase in temperature was markedly suppressed in PAFR-deficient mice. Acetyl-CoA:lyso-platelet-activating factor (PAF) acetyltransferase, which is involved in PAF biosynthesis, and the PAF receptor were predominantly localized in BAT macrophages, whereas brown adipocytes possessed the enzyme and functional PAF receptors. The stimulation of brown adipocytes by PAF induced the expression of ß3-AR mRNA and protein (1.5- and 1.9-fold, respectively), but not that of UCP1. These results indicate that obesity in PAFR-deficient mice resulted from impaired BAT activity and suggest that the antiobese function of PAF occurs through ß3-AR/UCP1 expression in BAT.


Assuntos
Adiposidade/fisiologia , Envelhecimento/fisiologia , Obesidade/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/deficiência , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/metabolismo , Tecido Adiposo Marrom/metabolismo , Adiposidade/genética , Animais , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Proteína Desacopladora 1
10.
Biochim Biophys Acta ; 1833(12): 2617-2627, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23816505

RESUMO

Claudin-4 is exclusively localized in the tight collecting ducts in the renal tubule. We examined what molecular mechanism is involved in the regulation of claudin-4 expression. In Madin-Darby canine kidney cells, hyperosmolarity increased the expression level of claudin-4 and the production of reactive oxygen species, which were inhibited by diphenyleneiodonium (DPI), an NADPH oxidase inhibitor, and manganese (III) tetrakis (4-benzoic acid)porphyrin (MnTBAP), a scavenger of H2O2. Both hyperosmolarity and H2O2 increased p-ERK1/2 and p-JNK, which were inhibited by U0126, a MEK inhibitor, and SP600125, a JNK inhibitor, respectively. Immunoprecipitation assay showed that hyperosmolarity increased the association of nuclear Sp1 with c-Jun, which was inhibited by U0126 and SP600125. In mouse inner medullary collecting duct cells and rat kidney slices, hyperosmolarity increased the expression level of claudin-4, which was inhibited by DPI, MnTBAP, U0126, and SP600125. Hyperosmolarity increased luciferase reporter activity of claudin-4, which was inhibited by U0126, SP600125, Sp1 siRNA, and c-Jun siRNA. The activity was inhibited by the mutation in the Sp1 binding site. Chromatin immunoprecipitation assay and avidin-biotin conjugated DNA assay showed that Sp1 and c-Jun are associated with the Sp1 binding site. These results suggest that hyperosmolarity increases nuclear Sp1/c-Jun complex and the association of the complex with the Sp1 binding site, resulting in the segment-specific expression of claudin-4 in the kidney.


Assuntos
Claudina-4/genética , Peróxido de Hidrogênio/metabolismo , Soluções Hipertônicas/farmacologia , NADPH Oxidases/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição Sp1/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Antracenos/farmacologia , Sequência de Bases , Butadienos/farmacologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Claudina-4/metabolismo , Cicloeximida/farmacologia , Cães , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sequestradores de Radicais Livres/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Células Madin Darby de Rim Canino , Modelos Biológicos , Dados de Sequência Molecular , Nitrilas/farmacologia , Concentração Osmolar , Regiões Promotoras Genéticas/genética , Ratos , Ratos Wistar
11.
Drug Metab Dispos ; 42(10): 1708-18, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25074870

RESUMO

The human pregnane X receptor (hPXR) is recognized as a xenobiotic-sensing nuclear receptor that transcriptionally regulates the gene expression of drug-metabolizing enzymes and transporters. Our study elucidates the mechanism by which the localization of hPXR is regulated through threonine-290. A phosphomimetic mutation at threonine-290 (T290D) retained hPXR in the cytoplasm of HepG2, HuH6, and SW480 cells in vitro and the mouse liver in vivo even after treatment with rifampicin, and a phosphodeficient mutation (T290A) translocated from the cytoplasm to the nucleus as the wild-type hPXR. The amount of the unphosphorylated wild-type yellow fluorescent protein-hPXR fusion protein but not the T290A mutant increased on Phos-tag gels in response to stimulations with rifampicin and cyclin-dependent kinase 2 inhibitor roscovitine, and a marked increase was observed in the unphosphorylated levels of the T290A mutant in nontreated cells. The Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN93 [2-[N-(2-hydroxyethyl)]-N-(4-methoxybenzenesulfonyl)]amino-N-(4-chlorocinnamyl)-N-methylbenzylamine)] and transfection with anti-CaMKII small-interfering RNA (siRNA) enhanced the unphosphorylated levels of the wild-type protein. CaMKII directly phosphorylated the threonine-290 of hPXR, and the T290A mutant conferred resistance to CaMKII. The protein phosphatase (PP) inhibitor okadaic acid (100 nM) and transfection with anti-PP1 siRNA but not anti-PP2A siRNA led to reduced expression of CYP3A4 mRNA. After the rifampicin and roscovitine stimulations, PP1 was recruited to the wild-type hPXR but not the T290A mutant. These results suggest that phosphorylation at threonine-290 by CaMKII may impair the function of hPXR by repressing its translocation to the nucleus, and dephosphorylation by PP1 is necessary for the xenobiotic-dependent nuclear translocation of hPXR.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Proteína Fosfatase 1/metabolismo , Receptores de Esteroides/química , Receptores de Esteroides/metabolismo , Treonina/metabolismo , Animais , Benzilaminas/farmacologia , Linhagem Celular , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucuronosiltransferase/biossíntese , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Mutação , Ácido Okadáico/farmacologia , Fosforilação/efeitos dos fármacos , Receptor de Pregnano X , Transporte Proteico , Purinas/farmacologia , RNA Interferente Pequeno/farmacologia , Rifampina/farmacologia , Roscovitina , Sulfonamidas/farmacologia , Treonina/genética
12.
Biochim Biophys Acta ; 1823(6): 1110-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22546605

RESUMO

In human adenocarcinoma, claudin-2 expression is higher than that in normal lung tissue, but the regulatory mechanism of its expression has not been clarified. In human adenocarcinoma A549 cells, claudin-2 level time-dependently increased under the control conditions. In contrast, claudin-1 expression remained constant for 24h. The concentration of epidermal growth factor (EGF) in medium time-dependently increased, which was inhibited by matrix metalloproteinase (MMP) inhibitor II, an inhibitor of MMP-1, 3, 7, and 9. MMP inhibitor II decreased claudin-2 and phosphorylated ERK1/2 (p-ERK1/2) levels, which were recovered by EGF. Both claudin-2 and p-ERK1/2 levels were decreased by EGF neutralizing antibody, EGF receptor (EGFR) siRNA, AG1478, an inhibitor of EGFR, U0126, an inhibitor of MEK, and the exogenous expression of dominant negative-MEK. These results suggest that EGF is secreted from A549 cells by MMP and increases claudin-2 expression mediated via the activation of an EGFR/MEK/ERK pathway. The inhibition of the signaling pathway decreased phosphorylated c-Fos and nuclear c-Fos levels. The introduction of c-Fos siRNA decreased claudin-2 level without affecting claudin-1. The promoter activity of human claudin-2 was decreased by AG1478 and U0126. Furthermore, the activity was decreased by the deletion or mutation of the AP-1 binding site of claudin-2 promoter. Chromatin immunoprecipitation and avidin-biotin conjugated DNA assays showed that c-Fos binds to the AP-1 binding site. We suggest that a secreted EGF up-regulates the transcriptional activity of claudin-2 mediated by the activation of an EGFR/MEK/ERK/c-Fos pathway in A549 cells.


Assuntos
Adenocarcinoma/enzimologia , Adenocarcinoma/genética , Claudinas/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-fos/metabolismo , Adenocarcinoma de Pulmão , Sítios de Ligação , Butadienos/farmacologia , Linhagem Celular Tumoral , Claudinas/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inibidores , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Nitrilas/farmacologia , Regiões Promotoras Genéticas/genética , Inibidores de Proteases/farmacologia , Ligação Proteica/efeitos dos fármacos , Quinazolinas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fator de Transcrição AP-1/metabolismo , Tirfostinas
13.
Biochim Biophys Acta ; 1808(6): 1502-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21073857

RESUMO

Rapamycin, an inhibitor of mammalian target of rapamycin (mTOR), is used in treatments for transplantation and cancer. Rapamycin causes hypomagnesemia, although precisely how has not been examined. Here, we investigated the effect of rapamycin on the expression of transient receptor potential melastatin 6 (TRPM6), a Mg2+ channel. Rapamycin and LY-294002, an inhibitor of phosphatidilinositol-3 kinase (PI3K) located upstream of mTOR, inhibited epidermal growth factor (EGF)-induced expression of the TRPM6 protein without affecting TRPM7 expression in rat renal NRK-52E epithelial cells. Both rapamycin and LY-294002 decreased EGF-induced Mg2+ influx. U0126, a MEK inhibitor, inhibited EGF-induced increases in c-Fos, p-ERK, and TRPM6 levels. In contrast, neither rapamycin nor LY-294002 inhibited EGF-induced increases in p-ERK and c-Fos levels. EGF increased p-Akt level, an effect inhibited by LY-294002 and 1L-6-hydroxymethyl-chiro-inositol2-[(R)-2-O-methyl-3-O-octadecylcarbonate] (Akt inhibitor). Akt inhibitor decreased TRPM6 level similar to rapamycin and LY-294002. These results suggest that a PI3K/Akt/mTOR pathway is involved in the regulation of TRPM6 expression. Rapamycin inhibited the EGF-induced increase in TRPM6 mRNA but did not inhibit human TRPM6 promoter activity. In the presence of actinomycin D, a transcriptional inhibitor, rapamycin accelerated the decrease in TRPM6 mRNA. Rapamycin decreased the expression and activity of a luciferase linked with the 3'-untranslated region of human TRPM6 mRNA. These results suggest that TRPM6 expression is up-regulated by a PI3K/Akt/mTOR pathway and rapamycin reduces TRPM6 mRNA stability, resulting in a decrease in the reabsorption of Mg2+.


Assuntos
Células Epiteliais/efeitos dos fármacos , Estabilidade de RNA/efeitos dos fármacos , Sirolimo/farmacologia , Canais de Cátion TRPM/genética , Animais , Butadienos/farmacologia , Linhagem Celular , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Immunoblotting , Imunossupressores/farmacologia , Inositol/análogos & derivados , Inositol/farmacologia , Túbulos Renais/citologia , Morfolinas/farmacologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
14.
J Cell Biochem ; 113(2): 499-507, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21938738

RESUMO

Claudin-4 regulates ion permeability via a paracellular pathway in renal epithelial cells, but its other physiological functions have not been examined. We found that hyperosmotic stress increases claudin-4 expression in Madin-Darby canine kidney cells. Here, we examined whether claudin-4 affects cell motility, cell association, and the intracellular distribution of endogenous junctional proteins. Doxycycline-inducible expression of claudin-4 did not change endogenous levels of claudin-1, claudin-2, claudin-3, occludin, E-cadherin, and ZO-1. Claudin-4 overexpression increased cell association and decreased cell migration without affecting cell proliferation. Doxycycline did not change cell junctional protein levels, cell association or cell migration in mock-transfected cells. The insolubility of claudin-1 and -3 in Triton X-100 was increased by claudin-4 overexpression, but that of claudin-2, occludin, ZO-1, and E-cadherin was unchanged. Immunocytochemistry showed that claudin-4 overexpression increases the accumulation of claudin-1 and -3 in tight junctions (TJs). Furthermore, claudin-4 overexpression increased the association of claudin-4 with claudin-1 and -3. These results suggest that claudin-4 accumulates claudin-1 and -3 in TJs to enhance cell-cell contact in renal tubular epithelial cells.


Assuntos
Claudinas/metabolismo , Células Epiteliais/metabolismo , Rim/citologia , Junções Íntimas/metabolismo , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Movimento Celular , Claudina-1 , Claudina-3 , Claudina-4 , Claudinas/genética , Cães , Doxiciclina , Impedância Elétrica , Células Epiteliais/fisiologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas de Membrana/metabolismo , Pressão Osmótica , Permeabilidade , Fosfoproteínas/metabolismo , Ligação Proteica , Transporte Proteico , Solubilidade , Estresse Fisiológico , Proteína da Zônula de Oclusão-1
15.
Drug Metab Dispos ; 40(10): 2031-40, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22829544

RESUMO

Human UDP-glucuronosyltransferase (UGT) 1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds such as bilirubin. The present study shows how cyclin-dependent kinase (CDK) inhibitor roscovitine stimulated the expression of UGT1A1 in HepG2 cells. Pregnane X receptor (PXR)-mediated transactivation of UGT1A1 reporter gene was more prominently enhanced by roscovitine, compared with the basal-, constitutive androstane receptor (CAR)-, and aryl hydrocarbon receptor-mediated activities. We determined the regulatory mechanism of UGT1A1 expression through PXR's stimulation by roscovitine. Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Immunoprecipitation analysis revealed that the T290D but not T408D mutant protein remained in cytoplasm by forming a complex with heat shock protein 90 and cytoplasmic CAR retention protein, whereas treatment with proteasome inhibitor MG-132 accumulated the T408D mutant protein in cytoplasm. Transfection with anti-CDK2 small interfering RNA (siRNA) but not anti-CDK1 or CDK5 siRNA led to enhanced expression of UGT1A1. S350D yellow fluorescent protein-PXR fusion protein could translocate from cytoplasm to nucleus similar to the wild-type protein but was detected as an acetylated protein, whose binding with retinoid X receptor (RXR) and histone deacetylase was impaired. Cotransfection with coactivator steroid receptor coactivator (SRC) 2 but not SRC-1 partly recovered its PXR activity. These results indicate that roscovitine stimulated the expression of UGT1A1 by inhibiting CDK2, which phosphorylated PXR at Ser350 to suppress binding with RXR and coactivator and maintain the acetylation of PXR protein.


Assuntos
Glucuronosiltransferase/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de Esteroides/metabolismo , Acetilação , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Receptor Constitutivo de Androstano , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/genética , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico , Células Hep G2 , Humanos , Imunoprecipitação , Chaperonas Moleculares , Mutação , Coativador 2 de Receptor Nuclear/metabolismo , Fosforilação , Receptor de Pregnano X , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico , Purinas/farmacologia , Interferência de RNA , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/efeitos dos fármacos , Receptores de Esteroides/genética , Receptores X de Retinoides/metabolismo , Rifampina/farmacologia , Roscovitina , Serina , Fatores de Tempo , Ativação Transcricional , Transfecção
16.
Biochim Biophys Acta ; 1798(3): 415-21, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19914201

RESUMO

Claudin-16 is involved in the paracellular reabsorption of Mg(2+) in the thick ascending limb of Henle. Little is known about the mechanism regulating the tight junctional localization of claudin-16. Here, we examined the effect of Mg(2+) deprivation on the distribution and function of claudin-16 using Madin-Darby canine kidney (MDCK) cells expressing FLAG-tagged claudin-16. Mg(2+) deprivation inhibited the localization of claudin-16 at tight junctions, but did not affect the localization of other claudins. Re-addition of Mg(2+) induced the tight junctional localization of claudin-16, which was inhibited by U0126, a MEK inhibitor. Transepithelial permeability to Mg(2+) was also inhibited by U0126. The phosphorylation of ERK was reduced by Mg(2+) deprivation, and recovered by re-addition of Mg(2+). These results suggest that the MEK/ERK-dependent phosphorylation of claudin-16 affects the tight junctional localization and function of claudin-16. Mg(2+) deprivation decreased the phosphothreonine levels of claudin-16. The phosphothreonine levels of T225A and T233A claudin-16 were decreased in the presence of Mg(2+) and these mutants were widely distributed in the plasma membrane. Furthermore, TER and transepithelial Mg(2+) permeability were decreased in the mutants. We suggest that the tight junctional localization of claudin-16 requires a physiological Mg(2+) concentration and the phosphorylation of threonine residues via a MEK/ERK-dependent pathway.


Assuntos
Espaço Extracelular/metabolismo , Magnésio/farmacologia , Proteínas de Membrana/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/enzimologia , Animais , Butadienos/farmacologia , Compartimento Celular/efeitos dos fármacos , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Claudinas , Cães , Impedância Elétrica , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Mutação/genética , Nitrilas/farmacologia , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotreonina/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Proteína da Zônula de Oclusão-1
17.
J Cell Physiol ; 226(9): 2448-56, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21660968

RESUMO

Epidermal growth factor (EGF) decreases the mRNA and protein levels of claudin-2 (CLDN2) in Madin-Darby canine kidney (MDCK) II cells. Here we examined whether EGF affects the stability and intracellular distribution of CLDN2 protein. EGF decreased surface and total levels of CLDN2, which was inhibited by U0126, a MEK inhibitor. CLDN2 was co-localized at the tight junctions (TJs) with ZO-1, a scaffolding protein. The fluorescence signal for CLDN2 disappeared on treatment with EGF, which was inhibited by U0126. EGF accelerated the decrease in CLDN2 in the presence of cycloheximide, a translation inhibitor, indicating that EGF reduces the stability of the protein. Chloroquine, a lysosomal protease inhibitor, blocked the EGF-induced decrease in CLDN2 protein and caused the co-localization of CLDN2 with Lamp-1, a marker of lysosome. Monodancylcadaverine, an inhibitor of endocytosis, and clathrin siRNA blocked the EGF-induced decrease in CLDN2 and the translocation of CLDN2 from the TJs to the lysosome. EGF increased the association of CLDN2 with clathrin and adaptin α which was inhibited by U0126. These results suggest that EGF accelerates clathrin-dependent endocytosis and lysosomal degradation of CLDN2 protein mediated by the activation of a MEK/ERK pathway.


Assuntos
Clatrina/metabolismo , Endocitose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Animais , Cadaverina/análogos & derivados , Cadaverina/farmacologia , Linhagem Celular , Cloroquina/farmacologia , Cães , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
18.
J Cell Physiol ; 226(6): 1471-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20717932

RESUMO

Migration of renal epithelial cells increases after renal tubular damage, but its mechanism has not been clarified in detail. Hyperosmotic stress increased a cellular injury concomitant with a decrease in mRNA and protein expression of claudin-2 in renal tubular epithelial Madin-Darby canine kidney cells. We hypothesized that claudin-2 is involved in the regulation of cell migration. To knockdown claudin-2 expression, we made the cells expressing doxycycline-inducible claudin-2 shRNA vector. Claudin-2 knockdown affected neither the endogenous expression levels of claudin-1, -3, -4, and -7 nor the Triton X-100 solubility of these claudins. Transepithelial electrical resistance was increased by claudin-2 knockdown without affecting permeability to FITC-dextran (4,000 Da). BrdU incorporation assay and cell counting revealed that cell proliferation and viability are unaffected by claudin-2 knockdown. In the wound-healing assay, the recovery rate of wound area was increased by claudin-2 knockdown. The mRNA expression and activity of matrix metalloproteinase-9 (MMP-9) were increased by claudin-2 knockdown. A selective MMP-9 inhibitor suppressed cell migration in the claudin-2 knockdown cells. Hyperosmotic stress increased the expression and activity of MMP-9, which were inhibited by claudin-2 overexpression. These results suggest that the decrease in claudin-2 expression enhances cell migration mediated by the increase in the expression and activity of MMP-9.


Assuntos
Movimento Celular , Claudinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Rim/patologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Claudinas/genética , Cães , Impedância Elétrica , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Pressão Osmótica/efeitos dos fármacos , Inibidores de Proteases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
19.
J Cell Biochem ; 112(12): 3563-72, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21780161

RESUMO

Lack of magnesium suppresses cell growth, but the molecular mechanism is not examined in detail. We examined the effect of extracellular magnesium deficiency on cell cycle progression and the expression of cell cycle regulators in renal epithelial NRK-52E cells. In synchronized cells caused by serum-starved method, over 80% cells were distributed in G1 phase. Cell proliferation and percentage of the cells in S phase in the presence of MgCl(2) were higher than those in the absence of MgCl(2) , suggesting that magnesium is involved in the cell cycle progression from G1 to S phase. After serum addition, the expression levels of p21(Cip1) and p27(Kip1) in the absence of MgCl(2) were higher than those in the presence of MgCl(2) . The exogenous expression of p21(Cip1) or p27(Kip1) increased the percentage in G1 phase, whereas it decreased that in S phase. The mRNA levels and promoter activities of p21(Cip1) and p27(Kip1) in the absence of MgCl(2) were higher than those in the presence of MgCl(2) . The phosphorylated p53 (p-p53) level was decreased by MgCl(2) addition. Pifithrin-α, a p53 inhibitor, decreased the p-p53, p21(Cip1) and p27(Kip1) levels, and the percentage in G1 phase in the absence of MgCl(2) . Rotenone, a mitochondrial respiratory inhibitor, decreased ATP content and increased the p-p53 level in the presence of MgCl(2) . Together, lack of magnesium may increase p21(Cip1) and p27(Kip1) levels mediated by the decrease in ATP content and the activation of p53, resulting in the suppression of cell cycle progression from G1 to S phase in NRK-52E cells.


Assuntos
Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Rim/metabolismo , Magnésio/metabolismo , Transcrição Gênica/fisiologia , Sequência de Bases , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Primers do DNA , Citometria de Fluxo , Humanos , Rim/citologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética
20.
J Cell Physiol ; 222(3): 481-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19937979

RESUMO

Transient receptor potential melastatin 6 (TRPM6) channel is involved in the reabsorption of magnesium in the kidney. We recently found that TRPM6 expression is up-regulated by EGF, but the regulatory mechanism has not been clear. TRPM6 mRNA was endogenously expressed in HEK293 cells. TRPM6 mRNA expression was increased by EGF, which was inhibited by U0126, an MEK inhibitor. Promoter activity of human TRPM6 was observed in the TRPM6 5'-flanking region from -1,214 to -718. This promoter activity was enhanced by EGF and inhibited by U0126. Three putative AP-1 binding sites were identified within the region of -1,214/-718. The mutation of the putative AP-1 binding site (-741/-736) completely inhibited the EGF-induced promoter activity. EGF increased p-ERK1/2, c-Fos, c-Jun, and p-c-Jun levels, which were inhibited by U0126. The introduction of c-Fos or c-Jun siRNA inhibited the EGF-induced promoter activity. A chromatin immunoprecipitation assay revealed that c-Fos and c-Jun bind to the AP-1 binding site within the region of -1,214/-718. These results suggest that EGF up-regulates TRPM6 mRNA expression mediate via the activation of ERK/AP-1-dependent pathway.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Células Epiteliais/metabolismo , Rim/metabolismo , Canais de Cátion TRPM/metabolismo , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Região 5'-Flanqueadora , Animais , Sequência de Bases , Sítios de Ligação , Butadienos/farmacologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Humanos , Rim/efeitos dos fármacos , Rim/enzimologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Mutação , Nitrilas/farmacologia , Fosforilação , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Canais de Cátion TRPM/genética , Fator de Transcrição AP-1/genética , Ativação Transcricional/efeitos dos fármacos , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA