Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Nutr ; 147(4): 482-498, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28250194

RESUMO

Background: Previously, we determined that heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) functions as an intracellular physiologic sensor of folate deficiency. In this model, l-homocysteine, which accumulates intracellularly in proportion to the extent of folate deficiency, covalently binds to and thereby activates homocysteinylated hnRNP-E1 to interact with folate receptor-α mRNA; this high-affinity interaction triggers the translational upregulation of cell surface folate receptors, which enables cells to optimize folate uptake from the external milieu. However, integral to this model is the need for ongoing generation of hnRNP-E1 to replenish homocysteinylated hnRNP-E1 that is degraded.Objective: We searched for an interrelated physiologic mechanism that could also maintain the steady-state concentration of hnRNP-E1 during prolonged folate deficiency.Methods: A novel RNA-protein interaction was functionally characterized by using molecular and biochemical approaches in vitro and in vivo.Results: l-homocysteine triggered a dose-dependent high-affinity interaction between hnRNP-E1 and a 25-nucleotide cis element within the 5'-untranslated region of hnRNP-E1 mRNA; this led to a proportionate increase in these RNA-protein complexes, and translation of hnRNP-E1 both in vitro and within placental cells. Targeted perturbation of this RNA-protein interaction either by specific 25-nucleotide antisense oligonucleotides or mutation within this cis element or by small interfering RNA to hnRNP-E1 mRNA significantly reduced cellular biosynthesis of hnRNP-E1. Conversely, transfection of hnRNP-E1 mutant proteins that mimicked homocysteinylated hnRNP-E1 stimulated both cellular hnRNP-E1 and folate receptor biosynthesis. In addition, ferrous sulfate heptahydrate [iron(II)], which also binds hnRNP-E1, significantly perturbed this l-homocysteine-triggered RNA-protein interaction in a dose-dependent manner. Finally, folate deficiency induced dual upregulation of hnRNP-E1 and folate receptors in cultured human cells and tumor xenografts, and more selectively in various fetal tissues of folate-deficient dams.Conclusions: This novel positive feedback loop amplifies hnRNP-E1 during prolonged folate deficiency and thereby maximizes upregulation of folate receptors in order to restore folate homeostasis toward normalcy in placental cells. It will also functionally impact several other mRNAs of the nutrition-sensitive, folate-responsive posttranscriptional RNA operon that is orchestrated by homocysteinylated hnRNP-E1.


Assuntos
Receptor 2 de Folato/metabolismo , Deficiência de Ácido Fólico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Placenta/citologia , Regulação para Cima/efeitos dos fármacos , Animais , Linhagem Celular , Proteínas de Ligação a DNA , Feminino , Receptor 2 de Folato/genética , Ácido Fólico/farmacologia , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Neoplasias do Colo do Útero/metabolismo
2.
Reprod Toxicol ; 53: 131-40, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25929818

RESUMO

The mouse Embryonic Stem cell Test (EST) using cardiomyocyte differentiation is a promising in vitro assay for detecting potential embryotoxicity; however, the addition of another differentiation endpoint, such as osteoblasts, may improve the predictive value of the test. A number of variables such as culture conditions and starting cell number were investigated. A 14 day direct plating method of D3 mouse embryonic stem cells (mESCs) was used to test the predictivity of osteoblast differentiation as an endpoint in the EST. Twelve compounds were tested using the prediction model developed in the ECVAM validation study. Eight of the compounds selected from the EST validation study served as model compounds; four additional compounds known to produce skeletal defects were also tested. Our results indicate comparable chemical classification between the validated cardiomyocyte endpoint and the osteoblast endpoint. These results suggest that differentiation to osteoblasts may provide confirmatory information in predicting embryotoxicity.


Assuntos
Osteoblastos/efeitos dos fármacos , Teratogênicos/toxicidade , Fosfatase Alcalina/metabolismo , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Hep G2 , Humanos , Camundongos , Células-Tronco Embrionárias Murinas , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Reprodutibilidade dos Testes
3.
J Biol Chem ; 286(45): 39100-15, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21930702

RESUMO

The mechanism underlying the sensing of varying degrees of physiological folate deficiency, prior to adaptive optimization of cellular folate uptake through the translational up-regulation of folate receptors (FR) is unclear. Because homocysteine, which accumulates intracellularly during folate deficiency, stimulated interactions between heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) and an 18-base FR-α mRNA cis-element that led to increased FR biosynthesis and net up-regulation of FR at cell surfaces, hnRNP-E1 was a plausible candidate sensor of folate deficiency. Accordingly, using purified components, we evaluated the physiological basis whereby L-homocysteine triggered these RNA-protein interactions to stimulate FR biosynthesis. L-homocysteine induced a concentration-dependent increase in RNA-protein binding affinity throughout the range of physiological folate deficiency, which correlated with a proportionate increase in translation of FR in vitro and in cultured human cells. Targeted reduction of newly synthesized hnRNP-E1 proteins by siRNA to hnRNP-E1 mRNA reduced both constitutive and L-homocysteine-induced rates of FR biosynthesis. Furthermore, L-homocysteine covalently bound hnRNP-E1 via multiple protein-cysteine-S-S-homocysteine mixed disulfide bonds within K-homology domains known to interact with mRNA. These data suggest that a concentration-dependent, sequential disruption of critical cysteine-S-S-cysteine bonds by covalently bound L-homocysteine progressively unmasks an underlying RNA-binding pocket in hnRNP-E1 to optimize interaction with FR-α mRNA cis-element preparatory to FR up-regulation. Collectively, such data incriminate hnRNP-E1 as a physiologically relevant, sensitive, cellular sensor of folate deficiency. Because diverse mammalian and viral mRNAs also interact with this RNA-binding domain with functional consequences to their protein expression, homocysteinylated hnRNP-E1 also appears well positioned to orchestrate a novel, nutrition-sensitive (homocysteine-responsive), posttranscriptional RNA operon in folate-deficient cells.


Assuntos
Receptor 1 de Folato/biossíntese , Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Homocisteína/metabolismo , Ligação Proteica , Proteínas de Ligação a DNA , Dissulfetos/metabolismo , Receptor 1 de Folato/genética , Deficiência de Ácido Fólico/genética , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas/genética , Homocisteína/genética , Humanos , Estrutura Terciária de Proteína , RNA Mensageiro , Proteínas de Ligação a RNA , Regulação para Cima
4.
OMICS ; 10(1): 1-14, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16584314

RESUMO

An integrated omics approach was undertaken in order to elucidate a systems biology level understanding of the acute hepatotoxcity of valproic acid (VPA). Metabonomics, proteomics and gene expression microarray platforms were employed in this systems biology study. CD-1 female pregnant mice were injected subcutaneously with 600 mg/kg VPA or vehicle control. Urine, serum, and liver tissue were collected at 6, 12, and 24 h after dosing. Principal component analysis (PCA) of the metabonomics data showed clustering of the dosed groups away from the controls for the urine samples. Looser clustering was seen in the other sample sets investigated. However, VPA administration resulted in altered glucose concentrations in urine samples at 12 and 24 h and in aqueous liver tissue extracts at 12 h after VPA administration. Proteomics studies identified two proteins, glycogen phosphorylase and amylo-1,6-glucosidase, which were increased in dosed animals relative to control. Both of these proteins are involved in converting glycogen to glucose. Examination of the expression of 20,000 liver genes did not reveal significantly altered expression at 6, 12, or 24 h after VPA exposure. The combined studies indicated a perturbation in the glycogenolysis pathway following administration of VPA.


Assuntos
Anticonvulsivantes/farmacologia , Fígado , Proteômica , Ácido Valproico/farmacologia , Animais , Anticonvulsivantes/metabolismo , Feminino , Perfilação da Expressão Gênica , Glucose/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/fisiologia , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/metabolismo , Gravidez , Ácido Valproico/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-15641086

RESUMO

BACKGROUND: Homocysteine, which increases in folate deficiency, can upregulate folate receptors (FR) at the translational level by increasing the interaction between a short cis-element in the 5'-untranslated region of FR-alpha mRNA and heterogeneous nuclear ribonucleoprotein-E1 (hnRNP-E1). Perturbation of this RNA-protein interaction on GD8.5 induces neural tube defects and neurocristopathies in mice. FR upregulation can also reduce cell proliferation independently of folate deficiency in some human cells. Accordingly, we tested the hypothesis that sustained murine maternal folate deficiency would negatively impact pregnancy outcomes, upregulate FR, and selectively reduce fetal cell proliferation. METHODS: Dams were fed chow with various levels of folic acid added for eight weeks before and throughout pregnancy. Following sacrifice on GD17, dams were compared for folate and homocysteine status as well as pregnancy outcomes. Fetuses from some groups were evaluated by specific biochemical, molecular, and immunohistochemical studies for FR, hnRNP-E1, and apoptosis. RESULTS: When compared to dams fed a folate-replete diet, those dams on a folate-depleted diet developed reduced red cell folates and hyperhomocysteinemia and an inverse dose-dependent upregulation of FR and hnRNP-E1 on GD17 without alterations in cell number in the majority of tissues. However, FR overexpression was accompanied by a significant reduction in the net number of cells in the midgut, lung, pons, tongue, and olfactory epithelium, and with premature differentiation in dorsal root ganglion cells and dysplasia of taste buds. By contrast, in the brain, spinal cord, diaphragm, and primordium of follicles of vibrissae, there was less FR expression, which accompanied a net reduction in number of cells and architectural anomalies. Subtle "immunohistochemical footprints" of apoptosis on GD17 fetuses corresponded with net cell loss in the lung and olfactory epithelium. Upregulation of FR could be explained by a homocysteine-induced RNA-protein interaction in folate-depleted fetuses that led to a proportionate increase in murine FR biosynthesis. CONCLUSIONS: Maternal folate deficiency results in selective upregulation of FR and hnRNP-E1 associated with multiple aberrations in fetal tissues that include increased cell loss, architectural anomalies, and premature differentiation. The potential significance of these findings to explain the wide spectrum of folate-responsive birth defects in humans is discussed.


Assuntos
Proteínas de Transporte/metabolismo , Feto/patologia , Ácido Fólico/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Diferenciação Celular/fisiologia , Eletroforese em Gel de Poliacrilamida , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Receptores de Folato com Âncoras de GPI , Imuno-Histoquímica , Fígado/metabolismo , Camundongos , Gravidez , Regulação para Cima , Vitamina B 12/metabolismo
6.
Birth Defects Res A Clin Mol Teratol ; 67(7): 475-87, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14565618

RESUMO

BACKGROUND: Although folic acid decreases the incidence of neural tube defects (NTDs) in humans, the mechanism for this protection is unknown. We have employed antisense technology to alter expression of the gene for the folate receptor (folate binding protein-1 [Folbp1]) in mouse embryos cultured in vitro. METHODS: Embryos were explanted on day 8 of gestation and cultured for 44 hr. Several oligodeoxyribonucleotides designed to modulate the coding region or a regulatory sequence in the 5'-untranslated region of Folbp1 were microinjected into the amniotic sac of embryos at the beginning of the culture period. RESULTS: Two different antisense sequences to the 5' and 3' coding region in Folbp1 produced concentration-dependent increases in the number of embryos with NTDs. Coinjection of 5-methyltetrahydrofolate with these sequences decreased the frequency of abnormal embryos. A semi-quantitative RT-PCR technique used to measure the amount of Folbp1 mRNA in treated and control embryos confirmed that the mRNA level was decreased by treatment with the antisense sequences. An antisense oligodeoxyribonucleotide to a 17 base cis regulatory element also generated a concentration-dependent increase in the frequency of embryos with NTDs, and a decrease in the level of Folbp1 mRNA. CONCLUSIONS: These results demonstrate that alterations in expression of Folbp1 by perturbing either the coding sequence or a critical regulatory cis-element can play a role in NTDs.


Assuntos
Proteínas de Transporte/genética , Defeitos do Tubo Neural/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fases de Leitura Aberta/genética , Sequências Reguladoras de Ácido Nucleico/genética , Regiões 5' não Traduzidas/genética , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Feminino , Receptores de Folato com Âncoras de GPI , Camundongos , Microinjeções , Gravidez , RNA Mensageiro/análise , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tetra-Hidrofolatos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA