Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Genes Dev ; 23(8): 986-96, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19390091

RESUMO

Small Heterodimer Partner (SHP) inhibits activities of numerous transcription factors involved in diverse biological pathways. As an important metabolic regulator, SHP plays a key role in maintaining cholesterol and bile acid homeostasis by inhibiting cholesterol conversion to bile acids. While SHP gene induction by increased bile acids is well established, whether SHP activity is also modulated remains unknown. Here, we report surprising findings that SHP is a rapidly degraded protein via the ubiquitin-proteasomal pathway and that bile acids or bile acid-induced intestinal fibroblast growth factor 19 (FGF19) increases stability of hepatic SHP by inhibiting proteasomal degradation in an extracellular signal-regulated kinase (ERK)-dependent manner. SHP was ubiquitinated at Lys122 and Lys123, and mutation of these sites altered its stability and repression activity. Tandem mass spectrometry revealed that upon bile acid treatment, SHP was phosphorylated at Ser26, within an ERK motif in SHP, and mutation of this site dramatically abolished SHP stability. Surprisingly, SHP stability was abnormally elevated in ob/ob mice and diet-induced obese mice. These results demonstrate an important role for regulation of SHP stability in bile acid signaling in normal conditions, and that abnormal stabilization of SHP may be associated with metabolic disorders, including obesity and diabetes.


Assuntos
Ácidos e Sais Biliares/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Ubiquitinação , Animais , Linhagem Celular Tumoral , Ácido Quenodesoxicólico/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fármacos Gastrointestinais/farmacologia , Meia-Vida , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Lisina/metabolismo , Camundongos , Mutação , Fosforilação , Estabilidade Proteica/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos
2.
Biochem J ; 444(1): 27-38, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22335598

RESUMO

UV radiation-mediated photodamage to the skin has been implicated in premature aging and photoaging-related skin cancer and melanoma. Little is known about the cellular events that underlie premature senescence, or how to impede these events. In the present study we demonstrate that PPARδ (peroxisome-proliferator-activated receptor δ) regulates UVB-induced premature senescence of normal keratinocytes. Activation of PPARδ by GW501516, a specific ligand of PPARδ, significantly attenuated UVB-mediated generation of ROS (reactive oxygen species) and suppressed senescence of human keratinocytes. Ligand-activated PPARδ up-regulated the expression of PTEN (phosphatase and tensin homologue deleted on chromosome 10) and suppressed the PI3K (phosphatidylinositol 3-kinase)/Akt pathway. Concomitantly, translocation of Rac1 to the plasma membrane, which leads to the activation of NADPH oxidases and generation of ROS, was significantly attenuated. siRNA (small interfering RNA)-mediated knockdown of PTEN abrogated the effects of PPARδ on cellular senescence, on PI3K/Akt/Rac1 signalling and on generation of ROS in keratinocytes exposed to UVB. Finally, when HR-1 hairless mice were treated with GW501516 before exposure to UVB, the number of senescent cells in the skin was significantly reduced. Thus ligand-activated PPARδ confers resistance to UVB-induced cellular senescence by up-regulating PTEN and thereby modulating PI3K/Akt/Rac1 signalling to reduce ROS generation in keratinocytes.


Assuntos
Queratinócitos/efeitos da radiação , PTEN Fosfo-Hidrolase/fisiologia , Superóxidos/antagonistas & inibidores , Animais , Membrana Celular/metabolismo , Células Cultivadas , Senescência Celular , Criança , Ativação Enzimática , Humanos , Queratinócitos/fisiologia , Ligantes , Camundongos , Camundongos Pelados , PPAR delta/agonistas , PPAR delta/fisiologia , PTEN Fosfo-Hidrolase/biossíntese , Fosfatidilinositol 3-Quinases/fisiologia , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Pele/citologia , Pele/metabolismo , Pele/efeitos da radiação , Superóxidos/metabolismo , Tiazóis/farmacologia , Raios Ultravioleta , Regulação para Cima , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
J Biol Chem ; 286(19): 17133-43, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21372136

RESUMO

Binding of N-formyl-methionyl-leucyl-phenylalanine (fMLP) to its specific cell surface receptor, N-formyl peptide receptor (FPR), triggers different cascades of biochemical events, eventually leading to cellular activation. However, the physiological role of fMLP and FPR during differentiation of mesenchymal stem cells is unknown. In this study, we attempted to determine whether fMLP regulates differentiation of mesenchymal stem cells derived from bone marrow. Analysis by quantitative-PCR and flow cytometry showed significantly increased expression of FPR1, but not FPR2 and FPR3, during osteoblastic differentiation. fMLP, a specific ligand of FPR1, promotes osteoblastic commitment and suppresses adipogenic commitment under differentiation conditions. Remarkably, fMLP-stimulated osteogenesis is associated with increased expression of osteogenic markers and mineralization, which were blocked by cyclosporine H, a selective FPR1 antagonist. In addition, fMLP inhibited expression of peroxisome proliferator-activated receptor-γ1, a major regulator of adipocytic differentiation. fMLP-stimulated osteogenic differentiation was mediated via FPR1-phospholipase C/phospholipase D-Ca(2+)-calmodulin-dependent kinase II-ERK-CREB signaling pathways. Finally, fMLP promoted bone formation in zebrafish and rabbits, suggesting its physiological relevance in vivo. Collectively, our findings provide novel insight into the functional role of fMLP in bone biology, with important implications for its potential use as a therapeutic agent for treatment of bone-related disorders.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Mesenquimais/citologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Osteoblastos/citologia , Receptores de Formil Peptídeo/fisiologia , Adipócitos/citologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Diferenciação Celular , Ciclosporina/farmacologia , Humanos , PPAR gama/metabolismo , Fosfolipase D/metabolismo , Coelhos , Receptores de Formil Peptídeo/química , Transdução de Sinais , Fosfolipases Tipo C/metabolismo
4.
Exp Mol Med ; 38(1): 55-62, 2006 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-16520553

RESUMO

Elevated expression of protein casein kinase II (CKII) stimulated basal phospholipase D (PLD) activity as well as PMA-induced PLD activation in human U87 astroglioma cells. Moreover, CKII-selective inhibitor, emodin and apigenin suppressed PMA-induced PLD activation in a dose-dependent manner as well as basal PLD activity, suggesting the involvement of CKII in the activation of both PLD1 and PLD2. CKII was associated with PLD1 and PLD2 in co-transfection experiments. Furthermore, CKII induced serine/threonine phosphorylation of PLD2 in vivo, and the multiple regions of PLD2 were phosphorylated by CKII in vitro kinase assay using glutathione S-transferase-PLD2 fusion protein fragments. Elevated expression of CKII or PLD increased cell proliferation but pretreatment of cells with 1-butanol suppressed CKII-induced cell proliferation. These results suggest that CKII is involved in proliferation of U87 cells at least in part, through stimulation of PLD activity.


Assuntos
Astrocitoma/enzimologia , Caseína Quinase II/farmacologia , Fosfolipase D/metabolismo , 1-Butanol/farmacologia , Astrocitoma/metabolismo , Astrocitoma/patologia , Western Blotting , Caseína Quinase II/análise , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glutationa Transferase/metabolismo , Humanos , Cinética , Fosfolipase D/genética , Fosforilação/efeitos dos fármacos , Testes de Precipitina , Proteínas Recombinantes de Fusão/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
5.
Ann N Y Acad Sci ; 1091: 244-57, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17341619

RESUMO

Recent studies demonstrated that constitutive androstane receptor (CAR) inhibits ER-mediated transactivation of both endogenous and synthetic estrogen responsive promotor in Hep G2. Whereas steroid and xenobiotic receptor (SXR) but not peroxisome proliferator-activated receptor-gamma (PPAR-gamma) was also reported to repress estrogen receptor (ER) transactivation of the synthetic 4ERE in Hep G2, the effects of these xenobiotic nuclear receptors (XNRs) on the endogenous estrogen responsive promotor remain to be determined. Effects of CAR, SXR, and PPAR-gamma on ER transactivation were also examined in three different kinds of breast cancer cell lines. However, except in MCF-7, studies were limited either in single dose response (MDA-MB-231) or with CAR only (MCF-7-K3). And there is presently no report on the effects of CAR, SXR, and PPAR-gamma on ER-mediated transactivation in ovarian-derived CHO-S cells. Accordingly, this article further examined the effects of the endogenous vitellogenin B1 estrogen responsive promotor on the SXR- and PPAR-gamma-modulated ER transactivation in Hep G2, and either dose-dependent or single dose effects of SXR, PPAR-gamma, and CAR in two different breast cancer cell lines and the ovarian-derived cell line respectively, on the ER-mediated transactivation of the synthetic (4ERE)-tk-luciferase reporter. Consistent with the previous report, CAR significantly repressed ER-mediated transactivation of the endogenous vitellogenin B1 promotor in Hep G2 cells. However, contrary to the effects on the synthetic promotor, PPAR-gamma potentiated whereas SXR did not have any effects on the ER transactivation of the vitellogenin promotor in Hep G2. In the breast cancer cell line of MDA-MB-231 in which endogenous ER is known not to be expressed, CAR modestly stimulated ER transactivation of the synthetic 4ERE in a low dose whereas both SXR and PPAR-gamma did not have any effects in all doses examined (20-500 ng). And in both CHO-S and estrogen-independent breast cancer cell line, MCF-7-K3, none of the three xenobiotic receptors significantly influenced the ER-mediated 4ERE transactivation in all doses examined. XNRs modulate ER-mediated transactivation depending on the estrogen response elements (EREs) and estrogen target cell types.


Assuntos
Estrogênios/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Estrogênio/fisiologia , Elementos de Resposta/fisiologia , Ativação Transcricional/fisiologia , Xenobióticos , Animais , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Estrogênios/fisiologia , Genes Reporter , Humanos , Luciferases/genética , Luciferases/metabolismo , Vitelogeninas/genética , Xenobióticos/metabolismo , Xenobióticos/farmacologia , Proteínas de Xenopus/genética
6.
Ann N Y Acad Sci ; 1091: 258-69, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17341620

RESUMO

Previous studies reported that constitutive androstane receptor (CAR) does not transactivate phenobarbital responsive unit (PBRU)2C1luciferase reporter gene in COS cells in which endogenous CYP2B1 gene is not induced with PB. In order to understand molecular mechanism(s) whereby PBRU is transactivated, this article determined if the use of strong thymidine kinase (TK) promotor rather than the minimal CYP2C1 promotor, and hepatocyte nuclear factor-4 (HNF-4) can affect CAR-mediated transactivation of PBRU in the monkey kidney epithelial-derived COS-7 cells. To examine CAR-mediated transactivation, cultured COS-7 cells were transfected with CAR expression plasmid, pEGFP-mCAR1, and confirmed for high level of the protein expression. In COS-7 cells, TK promotor induced CAR-mediated PBRU transactivation in a dose-dependent manner. Whereas expression of HNF-4 slightly promoted PBRU transactivation with low amount of CAR transfected, it repressed PBRU transactivation in a dose-dependent manner with high amount of CAR. Consistent with the previous reports in Hep G2 cells, CAR transactivated PBRU2C1luciferase in a dose-dependent manner and this CAR-mediated transactivation required functional NR-1 and NF-1 sites. However, HNF-4 did not affect CAR-mediated PBRU transactivation in Hep G2 cells. These results suggest that proximal promotor and a trans-acting factor, HNF-4, can affect CAR-mediated transactivation of PBRU in COS-7 cells.


Assuntos
Citocromo P-450 CYP2B1/genética , Regulação da Expressão Gênica/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Fenobarbital/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Elementos de Resposta/fisiologia , Timidina Quinase/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Receptor Constitutivo de Androstano , Citocromo P-450 CYP2B1/biossíntese , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Rim , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Elementos de Resposta/efeitos dos fármacos , Timidina Quinase/fisiologia
7.
Ann N Y Acad Sci ; 1041: 470-80, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15956748

RESUMO

This study was performed to determine whether relaxin influences the growth of human adenocarcinoma (HeLa) cells of the cervix, which has been shown to contain relaxin-binding sites and proliferate by relaxin. Cells were maintained in Dulbecco's modified Eagle's medium with 5% fetal calf serum. Highly purified porcine relaxin was added at concentrations of 10(-11) to 10(-5) M and incubated for 44, 69, 94, and 118 h. Cell proliferation/cytotoxicity and endogenous nitric oxide production were determined using a colorimetric MTS assay and Griess assay, respectively. The results indicate that whereas relaxin promotes the growth rate of MCF-7 cells, which is in agreement with previous reports, at the doses and times of exposure used in this study relaxin does not significantly influence the number of viable HeLa cells. However, relaxin at nanomolar concentrations (10(-9) to 10(-8) M) promoted the growth of HeLa cells in the presence of both estrogen and progesterone to a small extent. In agreement with these results, relaxin did not affect nitric oxide production. We conclude that relaxin may have differential effects on the growth of two different cancer cell types, MCF-7 and HeLa cells.


Assuntos
Adenocarcinoma/patologia , Relaxina/farmacologia , Neoplasias do Colo do Útero/patologia , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Feminino , Células HeLa , Humanos
8.
Exp Mol Med ; 36(2): 103-9, 2004 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-15150437

RESUMO

Oxidative stress has been implicated in mediation of vascular disorders. In the presence of vanadate, H(2)O(2) induced tyrosine phosphorylation of PLD1, protein kinase C-alpha (PKC-alpha), and other unidentified proteins in rat vascular smooth muscle cells (VSMCs). Interestingly, PLD1 was found to be constitutively associated with PKC-alpha in VSMCs. Stimulation of the cells by H(2)O(2) and vanadate showed a concentration-dependent tyrosine phosphorylation of the proteins in PLD1 immunoprecipitates and activation of PLD. Pretreatment of the cells with the protein tyrosine kinase inhibitor, genistein resulted in a dose-dependent inhibition of H(2)O(2)-induced PLD activation. PKC inhibitor and down-regulation of PKC abolished H(2)O(2)-stimulated PLD activation. The cells stimulated by oxidative stress (H(2)O(2)) caused increased cell migration. This effect was prevented by the pretreatment of cells with tyrosine kinase inhibitors, PKC inhibitors, and 1-butanol, but not 3-butanol. Taken together, these results suggest that PLD might be involved in oxidative stress-induced migration of VSMCs, possibly via tyrosine phosphorylation and PKC activation.


Assuntos
Movimento Celular/fisiologia , Músculo Liso Vascular/fisiologia , Estresse Oxidativo , Fosfolipase D/metabolismo , Proteína Quinase C/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Genisteína/farmacologia , Peróxido de Hidrogênio/farmacologia , Músculo Liso Vascular/citologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Vanadatos/farmacologia , Doenças Vasculares/metabolismo
9.
Sci Rep ; 3: 1384, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23463338

RESUMO

Busulfan and cyclophosphamide (B/C)-treated mice exhibited a marked increase in apoptosis and a concomitant decrease in the ovarian weight. Histological and RT-PCR analysis indicate that the period of germ cell depletion in the B/C-treated ovaries coincides with decreased expression of genes Figla, Lhx8, Nobox, c-kit, and Sox3. However, depletion of the ovarian germ cells is mediated by autophagy-independent pathways that involve Fas/FasL-, TNF-, and/or p53-signalings. Treatment with B/C resulted in the cease of the reproductive function to produce their offspring during the 15(th) week post-treatment period in female mice. Furthermore, injection of the 3 × 10(6) GFP positive primordial follicles into the ovaries of the B/C treated mouse did not show apparent colonization of the transplanted follicles within the recipient ovaries. The present results suggest that B/C treatment is closely associated with an increased risk of premature ovarian failure.


Assuntos
Antineoplásicos/efeitos adversos , Folículo Ovariano/transplante , Ovário/efeitos dos fármacos , Ovário/cirurgia , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/genética , Biomarcadores , Bussulfano/administração & dosagem , Bussulfano/efeitos adversos , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Ovário/patologia , Óvulo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Reprodução/efeitos dos fármacos , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
10.
Arch Pharm Res ; 35(9): 1655-64, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23054723

RESUMO

This study examined the effect of rosiglitazone, an oral antidiabetic drug, on human ether-a-gogo-related gene (HERG) channels expressed in human embryonic kidney (HEK293) cells. Using the whole-cell patch-clamp technique, interaction between rosiglitazone and HERG in HEK293 cells was studied. Rosiglitazone inhibited HERG channels in a concentration-dependent manner, with an IC50 value of 18.8 µM and a Hill coefficient of 1.0. These effects were reversible after wash-out of the drug. The rosiglitazone-induced inhibition of HERG channels was voltagedependent, with a steep increase in inhibition over the voltage range of channel opening. However, inhibition was voltage-independent over the voltage range in which channels are fully activated. Rosiglitazone did not change the steady-state activation or inactivation curves or the activation or deactivation kinetics, implying that rosiglitazone blocks HERG channels predominantly in the open and inactivated state rather than in the closed state. The present study suggests that rosiglitazone blocks HERG channels by binding to activated and inactivated channels, and rosiglitazone use should thus be carefully monitored in patients with pre-existing QT prolongation.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Hipoglicemiantes/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Tiazolidinedionas/farmacologia , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Concentração Osmolar , Técnicas de Patch-Clamp , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Rosiglitazona
11.
Korean Circ J ; 42(4): 266-73, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22563340

RESUMO

BACKGROUND AND OBJECTIVES: The comparison of long-term clinical effects between Sirolimus-eluting stent (SES) and Paclitaxel-eluting stents (PES) for treatment of acute myocardial infarction (AMI) remains unclear. Seeking to clarify this issue, we performed a retrospective analysis to evaluate four-year clinical outcomes of SES compared to PES treated AMI patients. SUBJECTS AND METHODS: From January 2004 to August 2006, all patients with acute ST-segment elevation myocardial infarction and acute non-ST segment elevation myocardial infarction who underwent percutaneous coronary intervention (PCI) by implantation of either SES or PES were enrolled. The occurrences of cardiac and non-cardiac deaths, recurrent infarction, target vessel revascularization (TVR) and stent thrombosis were analyzed. The composite end points of these major adverse cardiac events (MACE) were also analyzed. RESULTS: During the study period, a total of 668 AMI patients had visited, of which 522 patients (299 with SES and 223 with PES) were enrolled. During the four-year clinical follow-up, both groups showed similar occurrences of non-cardiac death (14.6±2.2% vs. 18.3±3.0%, p=0.26); cardiac death (6.8±1.52% vs. 11.2±2.6%, p=0.39); re-infarction (3.3±1.1% vs. 6.4±1.8%, p=0.31); and stent thrombosis (3.2±1.1% vs. 5.4±1.7%, p=0.53). However, occurrences of TVR {4.0±1.2% vs. 10.0±3.0%, hazard ratio (HR)=0.498, 95% confidence interval (CI)=0.257-0.967, p=0.039} and MACE (19.4±2.5% vs. 29.4±3.5%, HR=0.645, 95% CI=0.443-0.940, p=0.021) were significantly lower in the SES population. CONCLUSION: In AMI patients treated with either SES or PES implantation, the former had a significantly lower risk of TVR and MACE during four-year clinical follow-up. Rates of death, cardiac death or recurrent infarction, and stent thrombosis were similar.

12.
Korean J Physiol Pharmacol ; 15(5): 291-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22128262

RESUMO

The effect of cyclosporin A (CsA), an immunosuppressant, on human ether-a-go-go-related gene (HERG) channel as it is expressed in human embryonic kidney cells was studied using a whole-cell, patch-clamp technique. CsA inhibited the HERG channel in a concentration-dependent manner, with an IC(50) value and a Hill coefficient of 3.17 µM and 0.89, respectively. Pretreatment with cypermethrine, a calcineurin inhibitor, had no effect on the CsA-induced inhibition of the HERG channel. The CsA-induced inhibition of HERG channels was voltage-dependent, with a steep increase over the voltage range of the channel opening. However, the inhibition exhibited voltage independence over the voltage range of fully activated channels. CsA blocked the HERG channels predominantly in the open and inactivated states rather than in the closed state. Results of the present study suggest that CsA acts directly on the HERG channel as an open-channel blocker, and it acts independently of its effect on calcineurin activity.

13.
Exp Mol Med ; 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20844341

RESUMO

Ahead of Print article withdrawn by publisher.

14.
Exp Mol Med ; 42(11): 731-8, 2010 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-20871212

RESUMO

The nuclear receptors, steroid and xenobiotic receptor (SXR) and constitutive androstane receptor (CAR) play important functions in mediating lipid and drug metabolism in the liver. The present study demonstrates modulatory actions of estrogen in transactivations of SXR-mediated liver X receptor response element (LXRE) and CAR-mediated phenobarbital response element (PBRU). When human estrogen receptor (hERα) and SXR were exogenously expressed, treatment with either rifampicin or corticosterone promoted significantly the SXR-mediated transactivation of LXRE reporter gene in HepG2. However, combined treatment with estrogen plus either rifampicin or corticosterone resulted in less than 50% of the mean values of the transactivation by rifampicin or corticosterone alone. Thus, it is suggested that estrogen may repress the SXR-mediated transactivation of LXRE via functional cross-talk between ER and SXR. The CAR-mediated transactivation of PBRU was stimulated by hERa in the absence of estrogen. However, the potentiation by CAR agonist, TCPOBOP, was significantly repressed by moxestrol in the presence of ER. Thus, ER may play both stimulatory and inhibitory roles in modulating CAR-mediated transactivation of PBRU depending on the presence of their ligands. In summary, this study demonstrates that estrogen modulates transcriptional activity of SXR and CAR in mediating transactivation of LXRE and PBRU, respectively, of the nuclear receptor target genes through functional cross-talk between ER and the corresponding nuclear receptors.


Assuntos
Estrogênios/metabolismo , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Ativação Transcricional/efeitos dos fármacos , Receptor Constitutivo de Androstano , Corticosterona/farmacologia , Etinilestradiol/análogos & derivados , Etinilestradiol/farmacologia , Células Hep G2 , Humanos , Receptores X do Fígado , Receptores Nucleares Órfãos/metabolismo , Fenobarbital/metabolismo , Receptor de Pregnano X , Piridinas/farmacologia , Receptor Cross-Talk , Receptores Citoplasmáticos e Nucleares/agonistas , Elementos de Resposta , Rifampina/farmacologia , Ativação Transcricional/fisiologia
15.
Exp Mol Med ; 42(8): 555-64, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20625243

RESUMO

Rebamipide a gastroprotective drug, is clinically used for the treatment of gastric ulcers and gastritis, but its actions on gastric cancer are not clearly understood. Phospholipase D (PLD) is overexpressed in various types of cancer tissues and has been implicated as a critical factor in inflammation and carcinogenesis. However, whether rebamipide is involved in the regulation of PLD in gastric cancer cells is not known. In this study, we showed that rebamipide significantly suppressed the expression of both PLD1 and PLD2 at a transcriptional level in AGS and MKN-1 gastric cancer cells. Downregulation of PLD expression by rebamipide inhibited its enzymatic activity. In addition, rebamipide inhibited the transactivation of nuclear factor kappa B (NFkappaB), which increased PLD1 expression. Rebamipide or PLD knockdown significantly suppressed the expression of genes involved in inflammation and proliferation and inhibited the proliferation of gastric cancer cells. In conclusion, rebamipide-induced downregulation of PLD may contribute to the inhibition of inflammation and proliferation in gastric cancer.


Assuntos
Alanina/análogos & derivados , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inflamação/enzimologia , Fosfolipase D/genética , Quinolonas/farmacologia , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/patologia , Alanina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/patologia , Isoenzimas/genética , Isoenzimas/metabolismo , NF-kappa B/metabolismo , Fosfolipase D/metabolismo , Regiões Promotoras Genéticas/genética , Neoplasias Gástricas/genética , Transcrição Gênica/efeitos dos fármacos
16.
J Biol Chem ; 277(29): 26356-63, 2002 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-12000748

RESUMO

Phenobarbital (PB) induction of CYP2B genes is mediated by translocation of the constitutively active androstane receptor (CAR) to the nucleus. Interaction of CAR with p160 coactivators and enhancement of CAR transactivation by the coactivators have been shown in cultured cells. In the present studies, the interaction of CAR with the p160 coactivator glucocorticoid receptor-interacting protein 1 (GRIP1) was examined in vitro and in vivo. Binding of GRIP1 to CAR was shown by glutathione S-transferase (GST) pull-down and affinity DNA binding. N- or C-terminal fragments of GRIP1 that contained the central receptor-interacting domain bound to GST-CAR, but the presence of ligand increased the binding to GST-CAR of only the fragments containing the C-terminal region. In gel shift analysis, binding to CAR was observed only with GRIP1 fragments containing the C-terminal region, and the binding was increased by a CAR agonist and decreased by a CAR antagonist. Expression of GRIP1 enhanced CAR-mediated transactivation in cultured hepatic-derived cells 2-3-fold. In hepatocytes transfected in vivo, expression of exogenous GRIP1 alone induced transactivation of the CYP2B1 PB-dependent enhancer 15-fold, whereas CAR expression alone resulted in only a 3-fold enhancement in untreated mice. Remarkably, CAR and GRIP1 together synergistically transactivated the enhancer about 150-fold, which is approximately equal to activation by PB treatment. In PB-treated mice, expression of exogenous CAR alone had little effect, expression of GRIP1 increased transactivation about 2-fold, and with CAR and GRIP, a 4-fold activation was observed. In untreated mice, expression of GRIP resulted in nuclear translocation of green fluorescent protein-CAR. These results strongly suggest that a p160 coactivator functions in CAR-mediated transactivation in vivo in response to PB treatment and that the synergistic activation of CAR by GRIP in untreated animals results from both nuclear translocation and activation of CAR.


Assuntos
Transporte Ativo do Núcleo Celular , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Animais , Células Cultivadas , Receptor Constitutivo de Androstano , DNA/metabolismo , Escherichia coli , Glutationa Transferase/metabolismo , Humanos , Ligantes , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Coativador 2 de Receptor Nuclear , Fenobarbital/farmacologia , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Transfecção
17.
J Biol Chem ; 277(37): 34626-33, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12114525

RESUMO

Estrogen receptor (ER) activity can be modulated by the action of other nuclear receptors. To study whether ER activity is altered by orphan nuclear receptors that mediate the cellular response to xenobiotics, cross-talk between ER and constitutive androstane receptor (CAR), steroid and xenobiotic receptor, or peroxisome proliferator-activated receptor gamma was examined in HepG2 cells. Of these receptors, CAR substantially inhibited ER-mediated transcriptional activity of the vitellogenin B1 promoter as well as a synthetic estrogen responsive element (ERE)-containing promoter. Treatment with an agonist of CAR, 1,4-bis-(2-(3,5-dichloropyridoxyl))benzene, potentiated CAR-mediated transcriptional repression. In contrast, an antagonist of CAR, androstenol, alleviated the repression effect. Although CAR interacted with the ER in solution, CAR did not interact with the ER bound to the ERE. CAR/retinoid X receptor bound to the ERE but with much lower affinity than ER. Incremental amounts of CAR elicited a progressive reduction of the ER activity induced by the p160 coactivator glucocorticoid receptor interacting protein 1 (GRIP-1). In turn, increasing amounts of GRIP-1 progressively reversed the depression of ER activity by CAR. An agonist or antagonist of CAR potentiated or alleviated, respectively, the CAR-mediated repression of the GRIP-1-enhanced ER activity, which is consistent with the ability of theses ligands to increase or decrease, respectively, the interaction of CAR with GRIP-1. A CAR mutant that did not interact with GRIP-1 did not inhibit ER-mediated transactivation. Our data demonstrate that xenobiotic nuclear receptor CAR antagonizes ER-mediated transcriptional activity by squelching limiting amounts of p160 coactivator and imply that xenobiotics may influence ER function of female reproductive physiology, cell differentiation, tumorigenesis, and lipid metabolism.


Assuntos
Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Estrogênio/fisiologia , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Receptor Constitutivo de Androstano , Histona Acetiltransferases , Humanos , Dados de Sequência Molecular , Coativador 1 de Receptor Nuclear , Coativador 2 de Receptor Nuclear , Receptores do Ácido Retinoico/fisiologia , Elementos de Resposta , Receptores X de Retinoides , Ativação Transcricional
18.
Eur J Biochem ; 271(17): 3470-80, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15317582

RESUMO

We show that epigallocatechin-3 gallate (EGCG), a major component of green tea, stimulates phospholipase D (PLD) activity in U87 human astroglioma cells. EGCG-induced PLD activation was abolished by the phospholipase C (PLC) inhibitor and a lipase inactive PLC-gamma1 mutant, which is dependent on intracellular or extracellular Ca(2+), with the possible involvement of Ca(2+)/calmodulin-dependent protein kinase II (CaM kinase II). EGCG induced translocation of PLC-gamma1 from the cytosol to the membrane and PLC-gamma1 interaction with PLD1. EGCG regulates the activity of PLD by modulating the redox state of the cells, and antioxidants reverse this effect. Moreover, EGCG-induced PLD activation was reduced by PKC inhibitors or down-regulation of PKC. Taken together, these results show that, in human astroglioma cells, EGCG regulates PLD activity via a signaling pathway involving changes in the redox state that stimulates a PLC-gamma1 [Ins(1,4,5)P(3)-Ca(2+)]-CaM kinase II-PLD pathway and a PLC-gamma1 (diacylglycerol)-PKC-PLD pathway.


Assuntos
Antineoplásicos/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Catequina/análogos & derivados , Catequina/metabolismo , Fosfolipase D/metabolismo , Proteína Quinase C/metabolismo , Fosfolipases Tipo C/metabolismo , Antioxidantes/metabolismo , Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Linhagem Celular Tumoral , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Humanos , Oxirredução , Fosfolipase C gama , Espécies Reativas de Oxigênio/metabolismo , Chá/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa