Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Ginseng Res ; 43(4): 527-538, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31695561

RESUMO

BACKGROUND: Ginsenoside Rg1 was shown to exert ligand-independent activation of estrogen receptor (ER) via mitogen-activated protein kinase-mediated pathway. Our study aimed to delineate the mechanisms by which Rg1 activates the rapid ER signaling pathways. METHODS: ER-positive human breast cancer MCF-7 cells and ER-negative human embryonic kidney HEK293 cells were treated with Rg1 (10-12M, 10-8M), 17ß-estradiol (10-8M), or vehicle. Immunoprecipitation was conducted to investigate the interactions between signaling protein and ER in MCF-7 cells. To determine the roles of these signaling proteins in the actions of Rg1, small interfering RNA or their inhibitors were applied. RESULTS: Rg1 rapidly induced ERα translocation to plasma membrane via caveolin-1 and the formation of signaling complex involving linker protein (Shc), insulin-like growth factor-I receptor, modulator of nongenomic activity of ER (MNAR), ERα, and cellular nonreceptor tyrosine kinase (c-Src) in MCF-7 cells. The induction of extracellular signal-regulated protein kinase and mitogen-activated protein kinase kinase (MEK) phosphorylation in MCF-7 cells by Rg1 was suppressed by cotreatment with small interfering RNA against these signaling proteins. The stimulatory effects of Rg1 on MEK phosphorylation in these cells were suppressed by both PP2 (Src kinase inhibitor) and AG1478 [epidermal growth factor receptor (EGFR) inhibitor]. In addition, Rg1-induced estrogenic activities, EGFR and MEK phosphorylation in MCF-7 cells were abolished by cotreatment with G15 (G protein-coupled estrogen receptor-1 antagonist). The increase in intracellular cyclic AMP accumulation, but not Ca mobilization, in MCF-7 cells by Rg1 could be abolished by G15. CONCLUSION: Ginsenoside Rg1 exerted estrogenic actions by rapidly inducing the formation of ER containing signalosome in MCF-7 cells. Additionally, Rg1 could activate EGFR and c-Src ER-independently and exert estrogenic effects via rapid activation of membrane-associated ER and G protein-coupled estrogen receptor.

2.
Biochim Biophys Acta ; 1829(2): 231-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23168245

RESUMO

In the present study, a functional neuron restrictive silencer element (NRSE) was initially identified in the 5' flanking region (-83 to -67, relative to ATG) of human secretin receptor (hSCTR) gene by promoter assays coupled with scanning mutation analyses. The interaction of neuron restrictive silencer factor (NRSF) with this motif was later indicated via gel mobility shift and ChIP assays. The silencing activity of NRSF was confirmed by over-expression and also by shRNA knock-down of endogenous NRSF. These studies showed an inverse relationship between the expression levels of NRSF and hSCTR in the cells. As hSCTR gene was previously shown to be controlled by two GC-boxes which are regulated by the ratio of Sp1 to Sp3, in the present study, the functional interactions of NRSF and Sp proteins to regulate hSCTR gene was investigated. By co-immunoprecipitation assays, we found that NRSF could be co-precipitated with Sp1 as well as Sp3 in PANC-1 cells. Interestingly, co-expressions of these factors showed that NRSF could suppress Sp1-mediated, but not Sp3-mediated, transactivation of hSCTR. Taken together, we propose here that the down-regulatory effects of NRSF on hSCTR gene expression are mediated via its suppression on Sp1-mediated transactivation.


Assuntos
Receptores Acoplados a Proteínas G , Receptores dos Hormônios Gastrointestinais , Proteínas Repressoras , Fator de Transcrição Sp1/metabolismo , Animais , Regulação da Expressão Gênica , Humanos , Células PC12 , Regiões Promotoras Genéticas , Ligação Proteica , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3/genética , Fator de Transcrição Sp3/metabolismo , Ativação Transcricional
3.
PLoS One ; 7(6): e39913, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761926

RESUMO

Osmoregulation via maintenance of water and salt homeostasis is a vital process. In the brain, a functional secretin (SCT) and secretin receptor (SCTR) axis has recently been shown to mediate central actions of angiotensin II (ANGII), including initiation of water intake and stimulation of vasopressin (VP) expression and release. In this report, we provide evidence that estrogen-related receptor α (ERRα, NR3B1), a transcription factor mainly involved in metabolism, acts as an upstream activator of the SCT gene. In vitro studies using mouse hypothalamic cell line N-42 show that ERRα upregulates SCT promoter and gene expression. More importantly, knockdown of endogenous ERRα abolishes SCT promoter activation in response to hypertonic and ANGII stimulations. In mouse brain, ERRα coexpresses with SCT in various osmoregulatory brain regions, including the lamina terminalis and the paraventricular nucleus of the hypothalamus, and its expression is induced by hyperosmotic and ANGII treatments. Based on our data, we propose that both the upregulation of ERRα and/or the increased binding of ERRα to the mouse SCT promoter are two possible mechanisms for the elevated SCT expression upon hyperosmolality and central ANGII stimulation.


Assuntos
Angiotensina II/farmacologia , Receptor alfa de Estrogênio/fisiologia , Secretina/metabolismo , Regulação para Cima/fisiologia , Animais , Sequência de Bases , Imunoprecipitação da Cromatina , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Imuno-Histoquímica , Injeções Intraventriculares , Camundongos , Reação em Cadeia da Polimerase , Ratos
4.
ACS Nano ; 5(10): 7838-47, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21905691

RESUMO

Lanthanide-doped upconversion nanoparticles (UCNPs) are considered promising novel near-infrared (NIR) bioimaging agents with the characteristics of high contrast and high penetration depth. However, the interactions between charged UCNPs and mammalian cells have not been thoroughly studied, and the corresponding intracellular uptake pathways remain unclear. Herein, our research work involved the use of a hydrothermal method to synthesize polyvinylpyrrolidone-coated UCNPs (UCNP-PVP), and then a ligand exchange reaction was performed on UCNP-PVP, with the help of polyethylenimine (PEI) and poly(acrylic acid) (PAA), to generate UCNP-PEI and UCNP-PAA. These polymer-coated UCNPs demonstrated good dispersibility in aqueous medium, had the same elemental composition and crystal phase, shared similar TEM and dynamic light scattering (DLS) size distribution, and exhibited similar upconversion luminescence efficiency. However, the positively charged UCNP-PEI evinced greatly enhanced cellular uptake in comparison with its neutral or negative counterparts, as shown by multiphoton confocal microscopy and inductively coupled plasma mass spectrometry (ICP-MS) measurements. Meanwhile, we found that cationic UCNP-PEI can be effectively internalized mainly through the clathrin endocytic mechanism, as revealed by colocalization, chemical, and genetic inhibitor studies. This study elucidates the role of the surface polymer coatings in governing UCNP-cell interactions, and it is the first report on the endocytic mechanism of positively charged lanthanide-doped UCNPs. Furthermore, this study provides important guidance for the development of UCNPs as specific intracellular nanoprobes, allowing us to control the UCNP-cell interactions by tuning surface properties.


Assuntos
Érbio/química , Fluoretos/química , Imagem Molecular/métodos , Nanopartículas/química , Polímeros/química , Polímeros/metabolismo , Itérbio/química , Ítrio/química , Clorpromazina/farmacologia , Clatrina/antagonistas & inibidores , Clatrina/deficiência , Clatrina/genética , Vesículas Revestidas/efeitos dos fármacos , Vesículas Revestidas/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Ligantes , Medições Luminescentes , Nanopartículas/toxicidade , Polímeros/toxicidade , Propriedades de Superfície
5.
Kidney Int ; 79(3): 280-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20944548

RESUMO

Body fluid homeostasis is critical for the survival of living organisms and hence is tightly controlled. From initial studies on the effects of secretin (SCT) on renal water reabsorption in the 1940s and recent investigations of its role in cardiovascular and neuroendocrine functions, it has now become increasingly clear that this peptide is an integral component of the homeostatic processes that maintain body fluid balance. This review, containing some of our recent findings of centrally expressed SCT on water intake, focuses on the actions of SCT in influencing the physiological, neuroendocrine, and cardiovascular processes that subserve body fluid homeostasis.


Assuntos
Líquidos Corporais/metabolismo , Secretina/metabolismo , Equilíbrio Hidroeletrolítico , Animais , Sistema Cardiovascular/metabolismo , Ingestão de Líquidos , Comportamento de Ingestão de Líquido , Humanos , Rim/metabolismo , Sistemas Neurossecretores/metabolismo
6.
FASEB J ; 24(12): 5024-32, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20739612

RESUMO

Fluid balance is critical to life and hence is tightly controlled in the body. Angiotensin II (ANGII), one of the most important components of this regulatory system, is recognized as a dipsogenic hormone that stimulates vasopressin (VP) expression and release. However, detailed mechanisms regarding how ANGII brings about these changes are not fully understood. In the present study, we show initially that the osmoregulatory functions of secretin (SCT) in the brain are similar to those of ANGII in mice and, more important, we discovered the role of SCT as the link between ANGII and its downstream effects. This was substantiated by the use of two knockout mice, SCTR(-/-) and SCT(-/-), in which we show the absence of an intact SCT/secretin receptor (SCTR) axis resulted in an abolishment or much reduced ANGII osmoregulatory functions. By immunohistochemical staining and in situ hybridization, the proteins and transcripts of SCT and its receptor are found in the paraventricular nucleus (PVN) and lamina terminalis. We propose that SCT produced in the circumventricular organs is transported and released in the PVN to stimulate vasopressin expression and release. In summary, our findings identify SCT and SCTR as novel elements of the ANGII osmoregulatory pathway in maintaining fluid balance in the body.


Assuntos
Angiotensina II/farmacologia , Secretina/metabolismo , Secretina/farmacologia , Animais , Ingestão de Líquidos/efeitos dos fármacos , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Reação em Cadeia da Polimerase , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Secretina/genética , Vasopressinas/metabolismo , Equilíbrio Hidroeletrolítico/efeitos dos fármacos
7.
Int J Cancer ; 127(1): 43-54, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19904746

RESUMO

Secretin plays a key role in the regulation of normal cholangiocyte physiology via secretin receptor (SCTR). SCTR expression is upregulated during extrahepatic cholestasis induced by bile duct ligation and closely associated with cholangiocyte proliferative responses. Although well studied in normal cholangiocytes, the role of secretin and the expression of SCTR in the regulation of cholangiocarcinoma proliferation are unknown. In vitro, secretin (10(-7) M) displayed differential effects on normal cholangiocyte [H-69 and human intrahepatic biliary epithelial cell line (HIBEpiC)] and cholangiocarcinoma (Mz-ChA-1, HuH-28, TFK-1, SG231, CCLP1 and HuCC-T1) cell lines as such secretin is mitogenic for normal cholangiocytes and antiproliferative for cholangiocarcinoma. As expected in normal cholangiocytes (HIBEpiC), secretin increased intracellular cyclic adenosine monophosphate (cAMP) levels. However, the effect of secretin on intracellular cAMP levels was suppressed in Mz-ChA-1 cells. Secretin-stimulated intracellular cAMP levels in Mz-ChA-1 were restored by pretreatment with pertussis toxin, suggesting that the receptor coupled to Galpha(i) rather than Galpha(s). SCTR expression was found to be downregulated in 4 of the 6 cholangiocarcinoma cell lines evaluated and in human cholangiocarcinoma biopsy samples. In vivo, secretin significantly inhibited the tumor size and more than doubled tumor latency, which was associated with a decrease in proliferating cell nuclear antigen and an increase in cleaved-caspase 3 expression levels. Our results demonstrate that secretin and/or the modulation of SCTR expression might have potential as a therapeutic tool in the treatment of cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/patologia , AMP Cíclico/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Secretina/fisiologia , Transdução de Sinais/fisiologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Citometria de Fluxo , Imunofluorescência , Humanos , Reação em Cadeia da Polimerase
8.
Gene ; 443(1-2): 55-63, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19450668

RESUMO

RASSF1A, which is frequently found inactivated in human cancers, is revealed as a tumor suppressor gene in nasopharyngeal carcinoma (NPC). Using RASSF1A-expressing (NP69 and HK-1) and non-RASSF1A-expressing (C666-1) cell models, the transcriptional regulation of RASSF1A was studied. By deletion analysis of 3.1 kb of 5' flanking region, the core promoter of RASSF1A was identified in the region between -431 and -1 upstream of the translation start site. Sequence analysis of this core promoter revealed several putative transcription factor binding sties. Using NP69 cells and by block replacement mutagenesis, the presence of three functional GC-boxes were identified, to which by competitive and supershift electrophoretic mobility shift assays (EMSA), the in vitro bindings of Sp1 and Sp3 were suggested. The in vivo functions of Sp-proteins in regulating RASSF1A gene were then investigated by overexpression studies; among the tested Sp-proteins, Sp1 or Sp3, but not Sp4, was able to augment promoter activities. More interestingly, co-expression of Sp1 and Sp3 could synergistically enhance RASSF1A promoter function. UV irradiation induces oxidation stresses and hence is routinely used to investigate expressions of oncogenes and tumor suppressors. In this report, upon UV irradiation, the RASSF1A promoter activity and endogenous transcript levels were found to be reduced. By chromatin immunoprecipitation (ChIP) and EMSA, we demonstrated that the binding of Sp1 and Sp3 onto -431 to -202 were significantly reduced after UV irradiation. This UV-mediated effect on RASSF1A promoter, as shown by specific inhibitors that interrupt cellular pathways, is MEK1-, but not JNK-dependent. In summary, our data provided a simple model to explain the potential development of NPC, via silencing of the tumor suppressor RASSF1A by reduced bindings of activators Sp1 and Sp3 onto the GC-boxes in the core promoter of the gene.


Assuntos
Regulação para Baixo , Neoplasias Nasofaríngeas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Humanos , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 4/metabolismo , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Fatores de Transcrição Sp/metabolismo , Proteínas Supressoras de Tumor/genética , Raios Ultravioleta
9.
FEBS J ; 275(22): 5458-78, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18959737

RESUMO

As the key regulator of reproduction, gonadotropin-releasing hormone (GnRH) is released by neurons in the hypothalamus, and transported via the hypothalamo-hypophyseal portal circulation to the anterior pituitary to trigger gonadotropin release for gonadal steroidogenesis and gametogenesis. To achieve appropriate reproductive function, mammals have precise regulatory mechanisms; one of these is the control of GnRH synthesis and release. In the past, the scarcity of GnRH neurons and their widespread distribution in the brain hindered the study of GnRH gene expression. Until recently, the development of GnRH-expressing cell lines with properties similar to those of in vivo GnRH neurons and also transgenic mice facilitated GnRH gene regulation research. This minireview provides a summary of the molecular mechanisms for the control of GnRH-I and GnRH-II gene expression. These include basal transcription regulation, which involves essential cis-acting elements in the GnRH-I and GnRH-II promoters and interacting transcription factors, and also feedback control by gonadotropins and gonadal sex steroids. Other physiological stimuli, e.g. insulin and melatonin, will also be discussed.


Assuntos
Retroalimentação Fisiológica , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Animais , Hormônios Esteroides Gonadais , Gonadotropinas , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição , Transcrição Gênica
10.
BMC Physiol ; 7: 6, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17640386

RESUMO

BACKGROUND: Prolactin promotes proliferation of several cells. Prolactin receptor exists as two isoforms: long and short, which activate different transduction pathways including the Ca2+-dependent PKC-signaling. No information exists on the role of prolactin in the regulation of the growth of female cholangiocytes. The rationale for using cholangiocytes from female rats is based on the fact that women are preferentially affected by specific cholangiopathies including primary biliary cirrhosis. We propose to evaluate the role and mechanisms of action by which prolactin regulates the growth of female cholangiocytes. RESULTS: Normal cholangiocytes express both isoforms (long and short) of prolactin receptors, whose expression increased following BDL. The administration of prolactin to normal female rats increased cholangiocyte proliferation. In purified normal female cholangiocytes, prolactin stimulated cholangiocyte proliferation, which was associated with increased [Ca2+]i levels and PKCbeta-I phosphorylation but decreased PKCalpha phosphorylation. Administration of an anti-prolactin antibody to BDL female rats decreased cholangiocyte proliferation. Normal female cholangiocytes express and secrete prolactin, which was increased in BDL rats. The data show that prolactin stimulates normal cholangiocyte growth by an autocrine mechanism involving phosphorylation of PKCbeta-I and dephosphorylation of PKCalpha. CONCLUSION: We suggest that in female rats: (i) prolactin has a trophic effect on the growth of normal cholangiocytes by phosphorylation of PKCbeta-I and dephosphorylation of PKCalpha; and (iii) cholangiocytes express and secrete prolactin, which by an autocrine mechanism participate in regulation of cholangiocyte proliferation. Prolactin may be an important therapeutic approach for the management of cholangiopathies affecting female patients.


Assuntos
Ductos Biliares/citologia , Ductos Biliares/enzimologia , Sinalização do Cálcio/fisiologia , Proliferação de Células , Prolactina/metabolismo , Proteína Quinase C/metabolismo , Animais , Ductos Biliares/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Isoenzimas/metabolismo , Masculino , Fosforilação , Prolactina/farmacologia , Proteína Quinase C beta , Proteína Quinase C-alfa/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores da Prolactina/biossíntese
11.
Ann N Y Acad Sci ; 1070: 388-92, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16888197

RESUMO

Repressor element 1 (RE-1) is a negative, cis-acting regulatory element that interacts with the transcription factor RE-1-silencing transcription factor (REST). REST represses gene expression by two repressor domains that recruit other factors including mSin3 and CoREST. RE-1 has been identified in an increasing number of neuronal-specific genes, and recently, functional REST sites have also been discovered in VIP and PACAP genes. In the present article, we demonstrated for the first time that RE-1 sites are present in the 5' flanking regions of several secretin/PACAP/VIP genes by in silico analysis. This observation suggests that RE-1/REST is a common negative regulatory pathway of this peptide family.


Assuntos
Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Proteínas Repressoras/genética , Secretina/genética , Fatores de Transcrição/genética , Peptídeo Intestinal Vasoativo/genética , Animais , Biologia Computacional , Humanos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/classificação , Secretina/classificação , Peptídeo Intestinal Vasoativo/classificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...