Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Biol Chem ; 290(3): 1829-36, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25492871

RESUMO

Regulation of transcription elongation by positive transcription elongation factor b (P-TEFb) plays a central role in determining the state of cell activation, proliferation, and differentiation. In cells, P-TEFb exists in active and inactive forms. Its release from the inactive 7SK small nuclear ribonucleoprotein complex is a critical step for P-TEFb to activate transcription elongation. However, no good method exists to analyze this P-TEFb equilibrium in living cells. Only inaccurate and labor-intensive cell-free biochemical assays are currently available. In this study, we present the first experimental system to monitor P-TEFb activation in living cells. We created a bimolecular fluorescence complementation assay to detect interactions between P-TEFb and its substrate, the C-terminal domain of RNA polymerase II. When cells were treated with suberoylanilide hydroxamic acid, which releases P-TEFb from the 7SK small nuclear ribonucleoprotein, they turned green. Other known P-TEFb-releasing agents, including histone deacetylase inhibitors, bromodomain and extraterminal bromodomain inhibitors, and protein kinase C agonists, also scored positive in this assay. Finally, we identified 5'-azacytidine as a new P-TEFb-releasing agent. This release of P-TEFb correlated directly with activation of human HIV and HEXIM1 transcription. Thus, our visualization of P-TEFb activation by fluorescent complementation assay could be used to find new P-TEFb-releasing agents, compare different classes of agents, and assess their efficacy singly and/or in combination.


Assuntos
Fator B de Elongação Transcricional Positiva/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Apoptose , Azacitidina/química , Proteínas de Bactérias/química , Sistema Livre de Células , Teste de Complementação Genética , Glicerol/química , Células HEK293 , Células HeLa , Inibidores de Histona Desacetilases/química , Humanos , Ácidos Hidroxâmicos/química , Proteínas Luminescentes/química , Microscopia de Fluorescência , Plasmídeos/metabolismo , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Fatores de Tempo , Transcrição Gênica , Vorinostat
2.
Methods ; 66(2): 188-99, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23927839

RESUMO

Förster resonance energy transfer (FRET) between fluorescent proteins (FPs) provides insights into the proximities and orientations of FPs as surrogates of the biochemical interactions and structures of the factors to which the FPs are genetically fused. As powerful as FRET methods are, technical issues have impeded their broad adoption in the biologic sciences. One hurdle to accurate and reproducible FRET microscopy measurement stems from variable fluorescence backgrounds both within a field and between different fields. Those variations introduce errors into the precise quantification of fluorescence levels on which the quantitative accuracy of FRET measurement is highly dependent. This measurement error is particularly problematic for screening campaigns since minimal well-to-well variation is necessary to faithfully identify wells with altered values. High content screening depends also upon maximizing the numbers of cells imaged, which is best achieved by low magnification high throughput microscopy. But, low magnification introduces flat-field correction issues that degrade the accuracy of background correction to cause poor reproducibility in FRET measurement. For live cell imaging, fluorescence of cell culture media in the fluorescence collection channels for the FPs commonly used for FRET analysis is a high source of background error. These signal-to-noise problems are compounded by the desire to express proteins at biologically meaningful levels that may only be marginally above the strong fluorescence background. Here, techniques are presented that correct for background fluctuations. Accurate calculation of FRET is realized even from images in which a non-flat background is 10-fold higher than the signal.


Assuntos
Análise de Célula Única/métodos , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/química , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/química , Células HeLa , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/química , Microscopia de Fluorescência/métodos , Conformação Proteica , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Reprodutibilidade dos Testes , Razão Sinal-Ruído
3.
J Biol Chem ; 285(4): 2428-37, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19926790

RESUMO

An ability to measure the biochemical parameters and structures of protein complexes at defined locations within the cellular environment would improve our understanding of cellular function. We describe widely applicable, calibrated Förster resonance energy transfer methods that quantify structural and biochemical parameters for interaction of the human estrogen receptor alpha-isoform (ER alpha) with the receptor interacting domains (RIDs) of three cofactors (SRC1, SRC2, SRC3) in living cells. The interactions of ER alpha with all three SRC-RIDs, measured throughout the cell nucleus, transitioned from structurally similar, high affinity complexes containing two ER alphas at low free SRC-RID concentrations (<2 nm) to lower affinity complexes with an ER alpha monomer at higher SRC-RID concentrations (approximately 10 nm). The methods also showed that only a subpopulation of ER alpha was available to form complexes with the SRC-RIDs in the cell. These methods represent a template for extracting unprecedented details of the biochemistry and structure of any complex that is capable of being measured by Förster resonance energy transfer in the cellular environment.


Assuntos
Bioquímica/métodos , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Calibragem , Estradiol/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Estrogênios/farmacologia , Células HeLa , Humanos , Modelos Químicos , Coativador 1 de Receptor Nuclear/química , Coativador 1 de Receptor Nuclear/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Transfecção
4.
J Biomed Opt ; 13(3): 031202, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18601526

RESUMO

We have witnessed remarkable advances over the past decade in the application of optical techniques to visualize the genetically encoded fluorescent proteins (FPs) in living systems. The imaging of the FPs inside living cells has become an essential tool for studies of cell biology and physiology. FPs are now available that span the visible spectrum from deep blue to deep red, providing a wide choice of genetically encoded fluorescent markers. Furthermore, some FPs have been identified that have unusual characteristics that make them useful reporters of the dynamic behaviors of proteins inside cells. These additions to the FP toolbox are now being used for some very innovative live-cell imaging applications. Here, we will highlight the characteristics and uses of a few of these exceptional probes. Many different optical methods can be combined with the FPs from marine organisms to provide quantitative measurements in living systems.


Assuntos
Recuperação de Fluorescência Após Fotodegradação/tendências , Transferência Ressonante de Energia de Fluorescência/tendências , Proteínas Luminescentes/análise , Microscopia de Fluorescência/tendências , Mapeamento de Interação de Proteínas/tendências , Espectrometria de Fluorescência/tendências
5.
J Biomed Opt ; 13(3): 031207, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18601531

RESUMO

Forster resonance energy transfer (FRET) detection of protein interaction in living cells is commonly measured following the expression of interacting proteins genetically fused to the cyan (CFP) and yellow (YFP) derivatives of the Aequorea victoria fluorescent protein (FP). These FPs can dimerize at mM concentrations, which may introduce artifacts into the measurement of interaction between proteins that are fused with the FPs. Here, FRET analysis of the interaction between estrogen receptors (alpha isoform, ERalpha) labeled with "wild-type" CFP and YFP is compared with that of ERalpha labeled with "monomeric" A206K mutants of CFP and YFP. The intracellular equilibrium dissociation constant for the hormone-induced ERalpha-ERalpha interaction is similar for ERalpha labeled with wild-type or monomeric FPs. However, the measurement of energy transfer measured for ERalpha-ERalpha interaction in each cell is less consistent with the monomeric FPs. Thus, dimerization of the FPs does not affect the kinetics of ERalpha-ERalpha interaction but, when brought close together via ERalpha-ERalpha interaction, FP dimerization modestly improves FRET measurement.


Assuntos
Artefatos , Proteínas de Bactérias/metabolismo , Receptor alfa de Estrogênio/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência/métodos , Mapeamento de Interação de Proteínas/métodos , Receptores de Estrogênio/metabolismo , Proteínas de Bactérias/química , Dimerização , Proteínas de Fluorescência Verde/química , Células HeLa , Humanos , Proteínas Luminescentes/química , Sensibilidade e Especificidade
6.
Mol Endocrinol ; 21(1): 49-61, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17008385

RESUMO

Selective estrogen receptor modulators (SERMs) inhibit estrogen activation of the estrogen receptor (ER) in some tissues but activate ER in other tissues. These tissue-selective actions suggest that SERMs may be identified with tissue specificities that would improve the safety of breast cancer and hormone replacement therapies. The identification of an improved SERM would be aided by understanding the effects of each SERM on the structure and interactions of ER. To date, the inability to obtain structures of the full-length ER has limited our structural characterization of SERM action to their antiestrogenic effects on the isolated ER ligand binding domain. We studied the effects of estradiol and the clinically useful SERMs 4-hydroxytamoxifen and fulvestrant on the conformation of the full-length ERalpha dimer complex by comparing, in living human breast cancer cells, the amounts of energy transfer between fluorophores attached to different domains of ERalpha. Estradiol, 4-hydroxytamoxifen, and fulvestrant all promoted the rapid formation of ERalpha dimers with equivalent interaction kinetics. The amino- and carboxyl-terminal ERalpha domains both contain activation functions differentially affected by these ligands, but the positions of only the carboxyl termini differed upon binding with estradiol, 4-hydroxytamoxifen, or fulvestrant. The association of a specific ERalpha dimer conformation with the binding of ligands of different clinical effect will assist the identification of a SERM with optimal tissue-selective estrogenic and antiestrogenic activities. These studies also provide a roadmap for dissecting important structural and kinetic details for any protein complex from the quantitative analysis of energy transfer.


Assuntos
Receptor alfa de Estrogênio/química , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , DNA/química , Dimerização , Estradiol/análogos & derivados , Estradiol/química , Estrogênios/metabolismo , Transferência Ressonante de Energia de Fluorescência , Fulvestranto , Humanos , Cinética , Ligantes , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Tamoxifeno/análogos & derivados , Tamoxifeno/química
7.
Endocrinology ; 148(2): 538-47, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17095596

RESUMO

Novel estrogenic therapies are needed that ameliorate menopausal symptoms and have the bone-sparing effects of endogenous estrogens but do not promote breast or uterine cancer. Recent evidence suggests that selective activation of the estrogen receptor (ER)-beta subtype inhibits breast cancer cell proliferation. To establish whether ERbeta-selective ligands represent a viable approach to improve hormone therapy, we investigated whether the estrogenic activities present in an herbal extract, MF101, used to treat hot flashes, are ERbeta selective. MF101 promoted ERbeta, but not ERalpha, activation of an estrogen response element upstream of the luciferase reporter gene. MF101 also selectively regulates transcription of endogenous genes through ERbeta. The ERbeta selectivity was not due to differential binding because MF101 binds equally to ERalpha and ERbeta. Fluorescence resonance energy transfer and protease digestion studies showed that MF101 produces a different conformation in ERalpha from ERbeta when compared with the conformations produced by estradiol. The specific conformational change induced by MF101 allows ERbeta to bind to an estrogen response element and recruit coregulatory proteins that are required for gene activation. MF101 did not activate the ERalpha-regulated proliferative genes, c-myc and cyclin D1, or stimulate MCF-7 breast cancer cell proliferation or tumor formation in a mouse xenograft model. Our results demonstrate that herbal ERbeta-selective estrogens may be a safer alternative for hormone therapy than estrogens that nonselectively activate both ER subtypes.


Assuntos
Anemarrhena/química , Receptor beta de Estrogênio/genética , Extratos Vegetais/farmacologia , Ativação Transcricional/efeitos dos fármacos , Animais , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Carcinógenos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Dietilestilbestrol , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Conformação Molecular , Transplante de Neoplasias , Tamanho do Órgão/efeitos dos fármacos , Extratos Vegetais/metabolismo , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transplante Heterólogo , Útero/efeitos dos fármacos , Útero/patologia
8.
Mol Cell Biol ; 24(2): 675-86, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14701740

RESUMO

CCAAT/enhancer-binding protein alpha (C/EBPalpha) is one of the key transcription factors that mediate lineage specification and differentiation of multipotent myeloid progenitors into mature granulocytes. Although C/EBPalpha is known to induce granulopoiesis while suppressing monocyte differentiation, it is unclear how C/EBPalpha regulates this cell fate choice at the mechanistic level. Here we report that inducers of monocyte differentiation inhibit the alternate cell fate choice, that of granulopoiesis, through inhibition of C/EBPalpha. This inhibition is mediated by extracellular signal-regulated kinases 1 and/or 2 (ERK1/2), which interact with C/EBPalpha through an FXFP docking site and phosphorylate serine 21. As a consequence of C/EBPalpha phosphorylation, induction of granulocyte differentiation by C/EBPalpha or retinoic acid is inhibited. Our analysis of C/EBPalpha by fluorescent resonance energy transfer revealed that phosphorylation induces conformational changes in C/EBPalpha, increasing the distance between the amino termini of C/EBPalpha dimers. Thus, myeloid development is partly regulated by an ERK1/2-mediated change in the conformation of C/EBPalpha that favors monocyte differentiation by blocking granulopoiesis.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Granulócitos/citologia , Granulócitos/metabolismo , Leucopoese/fisiologia , Células 3T3-L1 , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Proteína alfa Estimuladora de Ligação a CCAAT/química , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Granulócitos/efeitos dos fármacos , Humanos , Células K562 , Leucopoese/efeitos dos fármacos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fosforilação , Serina/química , Tretinoína/farmacologia , Células U937
9.
Mol Endocrinol ; 19(7): 1675-86, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15761028

RESUMO

Hormones integrate the activities of their target cells through receptor-modulated cascades of protein interactions that ultimately lead to changes in cellular function. Understanding how the cell assembles these signaling protein complexes is critically important to unraveling disease processes, and to the design of therapeutic strategies. Recent advances in live-cell imaging technologies, combined with the use of genetically encoded fluorescent proteins, now allow the assembly of these signaling protein complexes to be tracked within the organized microenvironment of the living cell. Here, we review some of the recent developments in the application of imaging techniques to measure the dynamic behavior, colocalization, and spatial relationships between proteins in living cells. Where possible, we discuss the application of these different approaches in the context of hormone regulation of nuclear receptor localization, mobility, and interactions in different subcellular compartments. We discuss measurements that define the spatial relationships and dynamics between proteins in living cells including fluorescence colocalization, fluorescence recovery after photobleaching, fluorescence correlation spectroscopy, fluorescence resonance energy transfer microscopy, and fluorescence lifetime imaging microscopy. These live-cell imaging tools provide an important complement to biochemical and structural biology studies, extending the analysis of protein-protein interactions, protein conformational changes, and the behavior of signaling molecules to their natural environment within the intact cell.


Assuntos
Hormônios/fisiologia , Proteínas Luminescentes/análise , Microscopia de Fluorescência , Proteínas/análise , Espectrometria de Fluorescência/métodos , Animais , Células/química , Humanos , Receptores Citoplasmáticos e Nucleares/fisiologia
10.
Methods Mol Biol ; 1443: 79-104, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27246335

RESUMO

The ligand-regulated structure and biochemistry of nuclear receptor complexes are commonly determined by in vitro studies of isolated receptors, cofactors, and their fragments. However, in the living cell, the complexes that form are governed not just by the relative affinities of isolated cofactors for the receptor but also by the cell-specific sequestration or concentration of subsets of competing or cooperating cofactors, receptors, and other effectors into distinct subcellular domains and/or their temporary diversion into other cellular activities. Most methods developed to understand nuclear receptor function in the cellular environment involve the direct tagging of the nuclear receptor or its cofactors with fluorescent proteins (FPs) and the tracking of those FP-tagged factors by fluorescence microscopy. One of those approaches, Förster resonance energy transfer (FRET) microscopy, quantifies the transfer of energy from a higher energy "donor" FP to a lower energy "acceptor" FP attached to a single protein or to interacting proteins. The amount of FRET is influenced by the ligand-induced changes in the proximities and orientations of the FPs within the tagged nuclear receptor complexes, which is an indicator of the structure of the complexes, and by the kinetics of the interaction between FP-tagged factors. Here, we provide a guide for parsing information about the structure and biochemistry of nuclear receptor complexes from FRET measurements in living cells.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência/métodos , Receptores Citoplasmáticos e Nucleares/metabolismo , Células Cultivadas , Proteínas de Fluorescência Verde/metabolismo , Humanos , Ligantes , Ligação Proteica , Receptores Androgênicos/metabolismo , Receptores Citoplasmáticos e Nucleares/ultraestrutura
11.
PLoS One ; 11(3): e0151860, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26998755

RESUMO

Testosterone (T) and related androgens are performance enhancing drugs (PEDs) abused by some athletes to gain competitive advantage. To monitor unauthorized androgen abuse, doping control programs use mass spectrometry (MS) to detect androgens, synthetic anabolic-androgenic steroids (AASs) and their metabolites in an athlete's urine. AASs of unknown composition will not be detected by these procedures. Since AASs achieve their anabolic effects by activating the Androgen Receptor (AR), cell-based bioassays that measure the effect of a urine sample on AR activity are under investigation as complementary, pan-androgen detection methods. We evaluated an AR BioAssay as a monitor for androgen activity in urine pre-treated with glucuronidase, which releases T from the inactive T-glucuronide that predominates in urine. AR BioAssay activity levels were expressed as 'T-equivalent' concentrations by comparison to a T dose response curve. The T-equivalent concentrations of androgens in the urine of hypogonadal participants supplemented with T (in whom all androgenic activity should arise from T) were quantitatively identical to the T measurements conducted by MS at the UCLA Olympic Analytical Laboratory (0.96 ± 0.22). All 17 AASs studied were active in the AR BioAssay; other steroids were inactive. 12 metabolites of 10 commonly abused AASs, which are used for MS monitoring of AAS doping because of their prolonged presence in urine, had reduced or no AR BioAssay activity. Thus, the AR BioAssay can accurately and inexpensively monitor T, but its ability to monitor urinary AASs will be limited to a period immediately following doping in which the active AASs remain intact.


Assuntos
Anabolizantes/urina , Androgênios/urina , Substâncias para Melhoria do Desempenho/urina , Receptores Androgênicos/metabolismo , Esteroides/urina , Detecção do Abuso de Substâncias/métodos , Bioensaio , Humanos , Espectrometria de Massas , Testosterona/urina
12.
Mol Endocrinol ; 17(2): 209-22, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12554749

RESUMO

The pituitary-specific homeodomain protein Pit-1 cooperates with other transcription factors, including CCAAT/enhancer binding protein alpha (C/EBPalpha), in the regulation of pituitary lactotrope gene transcription. Here, we correlate cooperative activation of prolactin (PRL) gene transcription by Pit-1 and C/EBPalpha with changes in the subnuclear localization of these factors in living pituitary cells. Transiently expressed C/EBPalpha induced PRL gene transcription in pituitary GHFT1-5 cells, whereas the coexpression of Pit-1 and C/EBPalpha in HeLa cells demonstrated their cooperativity at the PRL promoter. Individually expressed Pit-1 or C/EBPalpha, fused to color variants of fluorescent proteins, occupied different subnuclear compartments in living pituitary cells. When coexpressed, Pit-1 recruited C/EBPalpha from regions of transcriptionally quiescent centromeric heterochromatin to the nuclear regions occupied by Pit-1. The homeodomain region of Pit-1 was necessary for the recruitment of C/EBPalpha. A point mutation in the Pit-1 homeodomain associated with the syndrome of combined pituitary hormone deficiency in humans also failed to recruit C/EBPalpha. This Pit-1 mutant functioned as a dominant inhibitor of PRL gene transcription and, instead of recruiting C/EBPalpha, was itself recruited by C/EBPalpha to centromeric heterochromatin. Together our results suggest that the intranuclear positioning of these factors determines whether they activate or silence PRL promoter activity.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Prolactina/genética , Fatores de Transcrição/genética , Transcrição Gênica , Transporte Ativo do Núcleo Celular/genética , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Células Cultivadas , Centrômero/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde , Heterocromatina/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Mutação , Hipófise/citologia , Hipófise/fisiologia , Prolactina/metabolismo , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição Pit-1 , Fatores de Transcrição/metabolismo
13.
Mol Endocrinol ; 17(3): 333-45, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12554785

RESUMO

The homeodomain protein Pit-1 cooperates with the basic-leucine zipper protein CCAAT/enhancer binding protein alpha (C/EBPalpha) to control pituitary-specific prolactin gene transcription. We previously observed that C/EBPalpha was concentrated in regions of centromeric heterochromatin in pituitary GHFT1-5 cells and that coexpressed Pit-1 redistributed C/EBPalpha to the subnuclear sites occupied by Pit-1. Here, we used fluorescence resonance energy transfer microscopy to show that when C/EBPalpha was recruited by Pit-1, the average distance separating the fluorophores labeling the proteins was less than 7 nm. A mutation in the Pit-1 homeodomain, or truncation of the C/EBPalpha transactivation domain disrupted the redistribution of C/EBPalpha by Pit-1. Fluorescence resonance energy transfer analysis revealed that the mutant Pit-1 still associated with C/EBPalpha, and the truncated C/EBPalpha still associated with Pit-1, but these interactions were preferentially localized in regions of centromeric heterochromatin. In contrast, a truncation in C/EBPalpha that prevented DNA binding also blocked its association with Pit-1, suggesting that the binding of C/EBPalpha to DNA is a critical first step in specifying its association with Pit-1. These findings indicated that the protein domains that specify the interaction of Pit-1 and C/EBPalpha are separable from the protein domains that direct the positioning of the associated proteins within the nucleus. The intimate association of Pit-1 and C/EBPalpha at certain sites within the living cell nucleus could foster their combinatorial activities in the regulation of pituitary-specific gene expression.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/fisiologia , Proteínas de Ligação a DNA/fisiologia , Hipófise/fisiologia , Fatores de Transcrição/fisiologia , Western Blotting , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Núcleo Celular/fisiologia , Cromatina/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Elementos Facilitadores Genéticos/genética , Elementos Facilitadores Genéticos/fisiologia , Recuperação de Fluorescência Após Fotodegradação , Transferência Ressonante de Energia de Fluorescência/métodos , Células HeLa , Proteínas de Homeodomínio/fisiologia , Humanos , Mutação , Hipófise/metabolismo , Fator de Transcrição Pit-1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia
14.
BMC Cell Biol ; 3: 6, 2002 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-11914124

RESUMO

BACKGROUND: A number of transcription factors coordinate differentiation by simultaneously regulating gene expression and cell proliferation. CCAAT/enhancer binding protein alpha (C/EBPalpha) is a basic/leucine zipper transcription factor that integrates transcription with proliferation to regulate the differentiation of tissues involved in energy balance. In the pituitary, C/EBPalpha regulates the transcription of a key metabolic regulator, growth hormone. RESULTS: We examined the consequences of C/EBPalpha expression on proliferation of the transformed, mouse GHFT1-5 pituitary progenitor cell line. In contrast to mature pituitary cells, GHFT1-5 cells do not contain C/EBPalpha. Ectopic expression of C/EBPalpha in the progenitor cells resulted in prolongation of both growth 1 (G1) and the DNA synthesis (S) phases of the cell cycle. Transcription activation domain 1 and 2 of C/EBPalpha were required for prolongation of G1, but not of S. Some transcriptionally inactive derivatives of C/EBPalpha remained competent for G1 and S phase prolongation. C/EBPalpha deleted of its leucine zipper dimerization functions was as effective as full-length C/EBPalpha in prolonging G1 and S. CONCLUSION: We found that C/EBPalpha utilizes mechanistically distinct activities to prolong the cell cycle in G1 and S in pituitary progenitor cells. G1 and S phase prolongation did not require that C/EBPalpha remained transcriptionally active or retained the ability to dimerize via the leucine zipper. G1, but not S, arrest required a domain overlapping with C/EBPalpha transcription activation functions 1 and 2. Separation of mechanisms governing proliferation and transcription permits C/EBPalpha to regulate gene expression independently of its effects on proliferation.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/fisiologia , DNA/biossíntese , Fase G1/fisiologia , Hipófise/metabolismo , Hipófise/fisiologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Transformada , Centrômero/genética , Fase G1/genética , Marcação de Genes , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Hipófise/citologia , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fase S/genética , Fase S/fisiologia , Células-Tronco/química , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Ativação Transcricional/genética , Ativação Transcricional/fisiologia
15.
PLoS One ; 9(9): e108693, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25259572

RESUMO

Insulin resistance, the diminished response of target tissues to insulin, is associated with the metabolic syndrome and a predisposition towards diabetes in a growing proportion of the worldwide population. Under insulin resistant states, the cellular response of the insulin signaling pathway is diminished and the body typically responds by increasing serum insulin concentrations to maintain insulin signaling. Some evidence indicates that the increased insulin concentration may itself further dampen insulin response. If so, insulin resistance would worsen as the level of circulating insulin increases during compensation, which could contribute to the transition of insulin resistance to more severe disease. Here, we investigated the consequences of excess insulin exposure to insulin receptor (IR) activity. Cells chronically exposed to insulin show a diminished the level of IR tyrosine and serine autophosphorylation below that observed after short-term insulin exposure. The diminished IR response did not originate with IR internalization since IR amounts at the cell membrane were similar after short- and long-term insulin incubation. Förster resonance energy transfer between fluorophores attached to the IR tyrosine kinase (TK) domain showed that a change in the TK domain occurred upon prolonged, but not short-term, insulin exposure. Even though the altered 'insulin refractory' IR TK FRET and IR autophosphorylation levels returned to baseline (non-stimulated) levels after wash-out of the original insulin stimulus, subsequent short-term exposure to insulin caused immediate re-establishment of the insulin-refractory levels. This suggests that some cell-based 'memory' of chronic hyperinsulinemic exposure acts directly at the IR. An improved understanding of that memory may help define interventions to reset the IR to full insulin responsiveness and impede the progression of insulin resistance to more severe disease states.


Assuntos
Hiperinsulinismo/metabolismo , Resistência à Insulina/fisiologia , Receptor de Insulina/metabolismo , Transdução de Sinais/fisiologia , Animais , Células CHO , Linhagem Celular Tumoral , Cricetulus , Insulina/metabolismo , Insulina/farmacologia , Fosforilação/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Ratos , Transdução de Sinais/efeitos dos fármacos
16.
PLoS One ; 8(5): e63286, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691010

RESUMO

The screening of large numbers of compounds or siRNAs is a mainstay of both academic and pharmaceutical research. Most screens test those interventions against a single biochemical or cellular output whereas recording multiple complementary outputs may be more biologically relevant. High throughput, multi-channel fluorescence microscopy permits multiple outputs to be quantified in specific cellular subcompartments. However, the number of distinct fluorescent outputs available remains limited. Here, we describe a cellular bar-code technology in which multiple cell-based assays are combined in one well after which each assay is distinguished by fluorescence microscopy. The technology uses the unique fluorescent properties of assay-specific markers comprised of distinct combinations of different 'red' fluorescent proteins sandwiched around a nuclear localization signal. The bar-code markers are excited by a common wavelength of light but distinguished ratiometrically by their differing relative fluorescence in two emission channels. Targeting the bar-code to cell nuclei enables individual cells expressing distinguishable markers to be readily separated by standard image analysis programs. We validated the method by showing that the unique responses of different cell-based assays to specific drugs are retained when three assays are co-plated and separated by the bar-code. Based upon those studies, we discuss a roadmap in which even more assays may be combined in a well. The ability to analyze multiple assays simultaneously will enable screens that better identify, characterize and distinguish hits according to multiple biologically or clinically relevant criteria. These capabilities also enable the re-creation of complex mixtures of cell types that is emerging as a central area of interest in many fields.


Assuntos
Núcleo Celular/metabolismo , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência/métodos , Imagem Molecular/métodos , Sequência de Aminoácidos , Biomarcadores/metabolismo , Proliferação de Células , Sobrevivência Celular , Técnicas de Cocultura , Células HeLa , Humanos , Proteínas Luminescentes/química , Dados de Sequência Molecular , Sinais de Localização Nuclear
17.
Methods Mol Biol ; 776: 147-66, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21796526

RESUMO

The androgen receptor (AR) is the central component of a dynamic conformational and interaction cascade initiated by androgenic hormones. AR function can be modified by cellular inputs not examined in test tube studies of AR action. Thus, there is a need to measure AR conformation and biochemistry directly within the cell where the intracellular locations, levels and availability of the hormone, AR, AR-interacting factors, DNA-binding sites, enzymes that modify those components of AR action, and factors that compete for the formation of functional AR-cofactor complexes may affect AR action. The dynamic nature of the AR functional cycle itself may introduce temporal fluctuations in factor status and location to affect AR output in the intact cell. This chapter focuses on the method of Förster resonance energy transfer which uniquely has the resolving power and ability to directly measure the conformation and biochemistry of AR signaling in living cells.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Animais , Calibragem , Técnicas de Cultura de Células , Humanos , Processamento de Imagem Assistida por Computador
18.
PLoS One ; 4(7): e6271, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19609440

RESUMO

Estrogens produce biological effects by interacting with two estrogen receptors, ERalpha and ERbeta. Drugs that selectively target ERalpha or ERbeta might be safer for conditions that have been traditionally treated with non-selective estrogens. Several synthetic and natural ERbeta-selective compounds have been identified. One class of ERbeta-selective agonists is represented by ERB-041 (WAY-202041) which binds to ERbeta much greater than ERalpha. A second class of ERbeta-selective agonists derived from plants include MF101, nyasol and liquiritigenin that bind similarly to both ERs, but only activate transcription with ERbeta. Diarylpropionitrile represents a third class of ERbeta-selective compounds because its selectivity is due to a combination of greater binding to ERbeta and transcriptional activity. However, it is unclear if these three classes of ERbeta-selective compounds produce similar biological activities. The goals of these studies were to determine the relative ERbeta selectivity and pattern of gene expression of these three classes of ERbeta-selective compounds compared to estradiol (E(2)), which is a non-selective ER agonist. U2OS cells stably transfected with ERalpha or ERbeta were treated with E(2) or the ERbeta-selective compounds for 6 h. Microarray data demonstrated that ERB-041, MF101 and liquiritigenin were the most ERbeta-selective agonists compared to estradiol, followed by nyasol and then diarylpropionitrile. FRET analysis showed that all compounds induced a similar conformation of ERbeta, which is consistent with the finding that most genes regulated by the ERbeta-selective compounds were similar to each other and E(2). However, there were some classes of genes differentially regulated by the ERbeta agonists and E(2). Two ERbeta-selective compounds, MF101 and liquiritigenin had cell type-specific effects as they regulated different genes in HeLa, Caco-2 and Ishikawa cell lines expressing ERbeta. Our gene profiling studies demonstrate that while most of the genes were commonly regulated by ERbeta-selective agonists and E(2), there were some genes regulated that were distinct from each other and E(2), suggesting that different ERbeta-selective agonists might produce distinct biological and clinical effects.


Assuntos
Receptor beta de Estrogênio/agonistas , Regulação da Expressão Gênica/efeitos dos fármacos , Western Blotting , Linhagem Celular , Estradiol/farmacologia , Transferência Ressonante de Energia de Fluorescência , Humanos , Lignanas , Nitrilas/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenóis/farmacologia , Propionatos/farmacologia , Transcrição Gênica/efeitos dos fármacos
19.
J Cell Sci ; 121(Pt 4): 437-49, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18230651

RESUMO

GAD65, the smaller isoform of the enzyme glutamic acid decarboxylase, synthesizes GABA for fine-tuning of inhibitory neurotransmission. GAD65 is synthesized as a soluble hydrophilic protein but undergoes a hydrophobic post-translational modification and becomes anchored to the cytosolic face of Golgi membranes. A second hydrophobic modification, palmitoylation of Cys30 and Cys45 in GAD65, is not required for the initial membrane anchoring but is crucial for post-Golgi trafficking of the protein to presynaptic clusters. The mechanism by which palmitoylation directs targeting of GAD65 through and out of the Golgi complex is unknown. Here, we show that prior to palmitoylation, GAD65 anchors to both ER and Golgi membranes. Palmitoylation, however, clears GAD65 from the ER-Golgi, targets it to the trans-Golgi network and then to a post-Golgi vesicular pathway. FRAP analyses of trafficking of GAD65-GFP reveal a rapid and a slow pool of protein replenishing the Golgi complex. The rapid pool represents non-palmitoylated hydrophobic GAD65-GFP, which exchanges rapidly between the cytosol and ER/Golgi membranes. The slow pool represents palmitoylation-competent GAD65-GFP, which replenishes the Golgi complex via a non-vesicular pathway and at a rate consistent with a depalmitoylation step. We propose that a depalmitoylation-repalmitoylation cycle serves to cycle GAD65 between Golgi and post-Golgi membranes and dynamically control levels of enzyme directed to the synapse.


Assuntos
Retículo Endoplasmático/metabolismo , Glutamato Descarboxilase/metabolismo , Complexo de Golgi/metabolismo , Lipoilação/fisiologia , Animais , Células Cultivadas , Recuperação de Fluorescência Após Fotodegradação , Glutamato Descarboxilase/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Membranas Intracelulares/metabolismo , Cinética , Lipoilação/genética , Modelos Biológicos , Transporte Proteico/fisiologia , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
20.
J Biol Chem ; 282(29): 20868-76, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17526489

RESUMO

Higher eukaryote genomes contain repetitive DNAs, often concentrated in transcriptionally inactive heterochromatin. Although repetitive DNAs are not typically considered as regulatory elements that directly affect transcription, they can contain binding sites for some transcription factors. Here, we demonstrate that binding of the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPalpha) to the mouse major alpha-satellite repetitive DNA sequesters C/EBPalpha in the transcriptionally inert pericentromeric heterochromatin. We find that this sequestration reduces the transcriptional capacity of C/EBPalpha. Functional sequestration of C/EBPalpha was demonstrated by experimentally reducing C/EBPalpha binding to the major alpha-satellite DNA, which elevated the concentration of C/EBPalpha in the non-heterochromatic subcompartment of the cell nucleus. The reduction in C/EBPalpha binding to alpha-satellite DNA was induced by the co-expression of the transcription factor Pit-1, which removes C/EBPalpha from the heterochromatic compartment, and by the introduction of an altered-specificity mutation into C/EBPalpha that reduces binding to alpha-satellite DNA but permits normal binding to sites in some gene promoters. In both cases the loss of alpha-satellite DNA binding coincided with an elevation in the binding of C/EBPalpha to a promoter and an increased transcriptional output from that promoter. Thus, the binding of C/EBPalpha to this highly repetitive DNA reduced the amount of C/EBPalpha available for binding to and regulation of this promoter. The functional sequestration of some transcription factors through binding to repetitive DNAs may represent an underappreciated mechanism controlling transcription output.


Assuntos
DNA/química , Sequências Repetitivas de Ácido Nucleico , Western Blotting , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Núcleo Celular/metabolismo , Centrômero/metabolismo , DNA Satélite/química , Heterocromatina/química , Mutação , Mutação Puntual , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Fator de Transcrição Pit-1/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA