Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nucleic Acids Res ; 50(15): e89, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-35641102

RESUMO

RNA polymerase II (RNAPII) is emerging as an important factor in DNA damage responses, but how it responds to genotoxic stress is not fully understood. We have developed a rapid and sensitive flow cytometry method to study chromatin binding of RNAPII in individual human cells through the cell cycle. Indicating enhanced transcription initiation at early timepoints, levels of RNAPII were increased at 15-30min after UV-induced DNA damage. This was particularly evident for the S5 phosphorylated form of RNAPII (pRNAPII S5), which is typically associated with promoter proximal pausing. Furthermore, degradation of pRNAPII S5 frequently occurs, as its levels on chromatin were strongly enhanced by the proteasome inhibitor MG132 with and without UV. Remarkably, inhibiting pause release with 5,6-dichloro-1-beta-ribo-furanosyl benzimidazole (DRB) further promoted UV-induced degradation of pRNAPII S5, suggesting enhanced initiation may lead to a phenomenon of 'promoter proximal crowding' resulting in premature termination via degradation of RNAPII. Moreover, pRNAPII S2 levels on chromatin were more stable in S phase of the cell cycle 2h after UV, indicating cell cycle specific effects. Altogether our results demonstrate a useful new method and suggest that degradation of promoter proximal RNAPII plays an unanticipated large role both during normal transcription and after UV.


Assuntos
Cromatina , Citometria de Fluxo/métodos , RNA Polimerase II , Cromatina/genética , Dano ao DNA , Humanos , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Análise de Célula Única , Transcrição Gênica
2.
Nucleic Acids Res ; 47(4): 1797-1813, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30541148

RESUMO

Ataxia telangiectasia mutated and Rad3-related (ATR) kinase is a key factor activated by DNA damage and replication stress. An alternative pathway for ATR activation has been proposed to occur via stalled RNA polymerase II (RNAPII). However, how RNAPII might signal to activate ATR remains unknown. Here, we show that ATR signaling is increased after depletion of the RNAPII phosphatase PNUTS-PP1, which dephosphorylates RNAPII in its carboxy-terminal domain (CTD). High ATR signaling was observed in the absence and presence of ionizing radiation, replication stress and even in G1, but did not correlate with DNA damage or RPA chromatin loading. R-loops were enhanced, but overexpression of EGFP-RNaseH1 only slightly reduced ATR signaling after PNUTS depletion. However, CDC73, which interacted with RNAPII in a phospho-CTD dependent manner, was required for the high ATR signaling, R-loop formation and for activation of the endogenous G2 checkpoint after depletion of PNUTS. In addition, ATR, RNAPII and CDC73 co-immunoprecipitated. Our results suggest a novel pathway involving RNAPII, CDC73 and PNUTS-PP1 in ATR signaling and give new insight into the diverse functions of ATR.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , RNA Polimerase II/genética , Estresse Fisiológico/genética , Proteínas Supressoras de Tumor/genética , Animais , Cromatina/genética , Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos da radiação , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Proteínas Nucleares/genética , Fosforilação/efeitos da radiação , Proteínas de Ligação a RNA/genética , Radiação Ionizante , Receptores de Neuropeptídeo Y/genética , Ribonuclease H/genética , Transdução de Sinais/efeitos da radiação , Estresse Fisiológico/efeitos da radiação
3.
Bio Protoc ; 13(8): e4659, 2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37113330

RESUMO

RNA polymerase II (RNAPII) transcribes DNA into mRNA and thereby plays a critical role in cellular protein production. In addition, RNAPII plays a central role in DNA damage responses. Measurements of RNAPII on chromatin may thus give insight into several essential processes in eukaryotic cells. During transcription, the C-terminal domain of RNAPII becomes post-translationally modified, and phosphorylation on serine 5 and serine 2 can be used as markers for the promoter proximal and productively elongating forms of RNAPII, respectively. Here, we provide a detailed protocol for the detection of chromatin-bound RNAPII and its serine 5- and serine 2-phosphorylated forms in individual human cells through the cell cycle. We have recently shown that this method can be used to study the effects of ultraviolet DNA damage on RNAPII chromatin binding and that it can even be used to reveal new knowledge about the transcription cycle itself. Other commonly used methods to study RNAPII chromatin binding include chromatin immunoprecipitation followed by sequencing or chromatin fractionation followed by western blotting. However, such methods are frequently based on lysates made from a large number of cells, which may mask population heterogeneity, e.g., due to cell cycle phase. With strengths such as single-cell analysis, speed of use, and accurate quantitative readouts, we envision that our flow cytometry method can be widely used as a complementary approach to sequencing-based methods to study effects of different stimuli and inhibitors on RNAPII-mediated transcription. Graphical overview.

4.
Int J Radiat Biol ; 99(6): 941-950, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-33877959

RESUMO

PURPOSE: Radiation-induced activation of cell cycle checkpoints have been of long-standing interest. The WEE1, CHK1 and ATR kinases are key factors in cell cycle checkpoint regulation and are essential for the S and G2 checkpoints. Here, we review the rationale for why inhibitors of WEE1, CHK1 and ATR could be beneficial in combination with radiation. CONCLUSIONS: Combined treatment with radiation and inhibitors of these kinases results in checkpoint abrogation and subsequent mitotic catastrophe. This might selectively radiosensitize tumor cells, as they often lack the p53-dependent G1 checkpoint and therefore rely more on the G2 checkpoint to repair DNA damage. Further affecting the repair of radiation damage, inhibition of WEE1, CHK1 or ATR also specifically suppresses the homologous recombination repair pathway. Moreover, inhibition of these kinases can induce massive replication stress during S phase of the cell cycle, likely contributing to eliminate radioresistant S phase cells. Intriguingly, recent findings suggest that cell cycle checkpoint inhibitors in combination with radiation can also enhance anti-tumor immune effects. Altogether, the expanding knowledge about the functional roles of WEE1, CHK1 and ATR inhibitors support that they are promising candidates for use in combination with radiation treatment.


Assuntos
Proteínas Tirosina Quinases , Radioterapia (Especialidade) , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Nucleares/metabolismo , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Dano ao DNA , Pontos de Checagem da Fase G2 do Ciclo Celular , Linhagem Celular Tumoral , Proteínas Mutadas de Ataxia Telangiectasia/genética
5.
EMBO Rep ; 11(11): 868-75, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20890310

RESUMO

The function of protein phosphatase 1 nuclear-targeting subunit (PNUTS)--one of the most abundant nuclear-targeting subunits of protein phosphatase 1 (PP1c)--remains largely uncharacterized. We show that PNUTS depletion by small interfering RNA activates a G2 checkpoint in unperturbed cells and prolongs G2 checkpoint and Chk1 activation after ionizing-radiation-induced DNA damage. Overexpression of PNUTS-enhanced green fluorescent protein (EGFP)--which is rapidly and transiently recruited at DNA damage sites--inhibits G2 arrest. Finally, γH2AX, p53-binding protein 1, replication protein A and Rad51 foci are present for a prolonged period and clonogenic survival is decreased in PNUTS-depleted cells after ionizing radiation treatment. We identify the PP1c regulatory subunit PNUTS as a new and integral component of the DNA damage response involved in DNA repair.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Núcleo Celular/enzimologia , Núcleo Celular/efeitos da radiação , Proteínas de Ligação a DNA/deficiência , Recuperação de Fluorescência Após Fotodegradação , Fase G2/efeitos da radiação , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Mitose/efeitos da radiação , Proteínas Nucleares/deficiência , RNA Interferente Pequeno/metabolismo , Radiação Ionizante , Proteínas Recombinantes de Fusão/metabolismo
6.
Mol Cell Oncol ; 8(3): 1910008, 2021 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-34027044

RESUMO

Conflicts between transcription and replication are a major source of replication stress. Our recent findings show that proper dephosphorylation of Serine 5 in the carboxy-terminal domain (CTD) of DNA-directed RNA polymerase II subunit RPB1 is needed to prevent such conflicts in human cells.

7.
Cell Rep ; 33(9): 108469, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33264625

RESUMO

Transcription-replication (T-R) conflicts cause replication stress and loss of genome integrity. However, the transcription-related processes that restrain such conflicts are poorly understood. Here, we demonstrate that the RNA polymerase II (RNAPII) C-terminal domain (CTD) phosphatase protein phosphatase 1 (PP1) nuclear targeting subunit (PNUTS)-PP1 inhibits replication stress. Depletion of PNUTS causes lower EdU uptake, S phase accumulation, and slower replication fork rates. In addition, the PNUTS binding partner WDR82 also promotes RNAPII-CTD dephosphorylation and suppresses replication stress. RNAPII has a longer residence time on chromatin after depletion of PNUTS or WDR82. Furthermore, the RNAPII residence time is greatly enhanced by proteasome inhibition in control cells but less so in PNUTS- or WDR82-depleted cells, indicating that PNUTS and WDR82 promote degradation of RNAPII on chromatin. Notably, reduced replication is dependent on transcription and the phospho-CTD binding protein CDC73 after depletion of PNUTS/WDR82. Altogether, our results suggest that RNAPII-CTD dephosphorylation is required for the continuous turnover of RNAPII on chromatin, thereby preventing T-R conflicts.


Assuntos
Cromatina/efeitos dos fármacos , Proteínas Cromossômicas não Histona/uso terapêutico , RNA Polimerase II/metabolismo , Proteínas Cromossômicas não Histona/farmacologia , Humanos , Transfecção
8.
Nat Biotechnol ; 20(5): 460-6, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11981558

RESUMO

We demonstrate here the functional reprogramming of a somatic cell using a nuclear and cytoplasmic extract derived from another somatic cell type. Reprogramming of 293T fibroblasts in an extract from primary human T cells or from a transformed T-cell line is evidenced by nuclear uptake and assembly of transcription factors, induction of activity of a chromatin remodeling complex, histone acetylation, and activation of lymphoid cell specific genes. Reprogrammed cells express T cell specific receptors and assemble the interleukin-2 receptor in response to T cell receptor CD3 (TCR CD3) complex stimulation. Reprogrammed primary skin fibroblasts also express T cell specific antigens. After exposure to a neuronal precursor extract, 293T fibroblasts express a neurofilament protein and extend neurite-like outgrowths. In vitro reprogramming of differentiated somatic cells creates possibilities for producing isogenic replacement cells for therapeutic applications.


Assuntos
Fibroblastos/metabolismo , Técnicas Genéticas , Linfócitos T/fisiologia , Biotecnologia/métodos , Complexo CD3/metabolismo , Diferenciação Celular , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citoplasma/metabolismo , Histonas/metabolismo , Humanos , Interleucina-2/metabolismo , Células Jurkat , Microscopia de Fluorescência , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transcrição Gênica
9.
Biochem J ; 390(Pt 3): 709-17, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15907195

RESUMO

PP1 (protein phosphatase-1) is a serine/threonine phosphatase involved in mitosis exit and chromosome decondensation. In the present study, we characterize the subcellular and subnuclear localization of PNUTS (PP1 nuclear targeting subunit), a nuclear regulatory subunit of PP1, and report a stimulatory role of PNUTS in the decondensation of prometaphase chromosomes in two in vitro systems. In interphase, PNUTS co-fractionates, together with a fraction of nuclear PP1, primarily with micrococcal nuclease-soluble chromatin. Immunofluorescence analysis shows that PNUTS is targeted to the reforming nuclei in telophase following the assembly of nuclear membranes and concomitantly with chromatin decondensation. In interphase cytosolic extract, ATP-dependent decondensation of prometaphase chromosomes is blocked by PP1-specific inhibitors. In contrast, a recombinant PNUTS(309-691) fragment accelerates chromosome decondensation. This decondensation-promoting activity requires the consensus RVXF PP1-binding motif of PNUTS, whereas a secondary, inhibitory PP1-binding site is dispensable. In a defined buffer system, PNUTS(309-691) also elicits decondensation in an exogenous PP1-dependent manner and, as in the cytosolic extract, a W401A (Thr401-->Ala) mutation that destroys PP1 binding abolishes this activity. The results illustrate an involvement of the PNUTS:PP1 holoenzyme in chromosome decondensation in vitro and argue that PNUTS functions as a PP1-targeting subunit in this process. We hypothesize that targeting of PNUTS to reforming nuclei in telophase may be a part of a signalling event promoting chromatin decondensation as cells re-enter interphase.


Assuntos
Cromossomos Humanos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Núcleo Celular/metabolismo , Células HeLa , Humanos , Interfase/fisiologia , Mitose/fisiologia , Fosfoproteínas Fosfatases/química , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 1 , Transporte Proteico
10.
Eur J Cell Biol ; 81(2): 69-76, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11893084

RESUMO

Follicles of the ileal Peyer's patch are sites of B cell proliferation and of diversification of the primary immunoglobulin repertoire in ruminants. We demonstrate here that 50-nm carbonic anhydrase-reactive particles released in the intercellular space in the follicle-associated epithelium of the ileal Peyer's patch of lambs contain DNA protected with a detergent-resistant membrane. We named these particles DiCAPs (DNA in carbonic anhydrase particles). DiCAPs can be purified from a suspension collected from ileal Peyer's patch follicles by sedimentation in a sucrose gradient. The DiCAP membrane is resistant to several ionic and non-ionic detergents alone, but can be disrupted by a combination of Triton X-100 and proteinase K. Differential nuclease treatment of purified DiCAPs indicates that they contain DNA. Digestion of DiCAP DNA with six-base pair restriction enzymes produces smears, suggesting that individual DiCAPs contain unique sequences. Nonetheless, the size of DiCAP DNA is smaller (approximately 16 kb) than that of lamb genomic DNA. Polymerase chain reaction and sequence analysis of DiCAP DNA reveals the presence of light and heavy chain variable genes as well as housekeeping genes. The data demonstrate the presence of DNA in these extracellular particles, and suggest a role of DiCAPs in transfer of DNA between cells within the ileal Peyer's patch. This raises the possibility of a novel form of communication between cells mediated by nucleic acids.


Assuntos
Anidrases Carbônicas/metabolismo , DNA/metabolismo , Espaço Extracelular/metabolismo , Íleo/metabolismo , Nódulos Linfáticos Agregados/metabolismo , Ovinos/metabolismo , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Sequência de Bases/genética , Anidrases Carbônicas/imunologia , Anidrases Carbônicas/ultraestrutura , Membrana Celular/imunologia , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , DNA/genética , DNA/imunologia , Espaço Extracelular/imunologia , Genes/genética , Genes/imunologia , Íleo/imunologia , Íleo/ultraestrutura , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Substâncias Macromoleculares , Membranas/imunologia , Membranas/metabolismo , Membranas/ultraestrutura , Microscopia Eletrônica , Dados de Sequência Molecular , Tamanho da Partícula , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/ultraestrutura , Ovinos/anatomia & histologia , Ovinos/imunologia
11.
Cell Cycle ; 10(9): 1356-62, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21451260

RESUMO

In response to DNA damaging agents and endogenous DNA lesions, human cells activate signaling cascades and repair mechanisms to help maintain genomic integrity. Phosphorylation plays a major role in DNA damage signaling, and the role of Ser/Thr kinases, including ATM, ATR, CHK1, CHK2 and DNA-PK, is particularly well documented. While these kinases have taken the center stage in DNA damage signaling until now, a role for Ser/Thr phosphatases is emerging, including Protein Phosphatase 1 (PP1). PP1 substrate specificity is regulated by its binding to a large number of different targeting subunits, and several of these have recently been identified as regulators of DNA damage responses. Here we review recent progress regarding the involvement of PP1 and its binding partners in DNA damage signaling.


Assuntos
Dano ao DNA/fisiologia , Proteína Fosfatase 1/metabolismo , Transdução de Sinais/fisiologia , Animais , Dano ao DNA/genética , Humanos , Fosforilação/genética , Fosforilação/fisiologia , Proteína Fosfatase 1/genética , Transdução de Sinais/genética
12.
Biochemistry ; 45(50): 14980-9, 2006 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-17154535

RESUMO

A-Kinase anchoring proteins (AKAPs) control the subcellular localization and temporal specificity of protein phosphorylation mediated by cAMP-dependent protein kinase. AKAP149 (AKAP1) is found in mitochondria and in the endoplasmic reticulum-nuclear envelope network where it anchors protein kinases, phosphatases, and a phosphodiesterase. AKAP149 harbors in its COOH-terminal part one KH and one Tudor domain, both known to be involved in RNA binding. We investigated the properties of the COOH-terminal domain of AKAP149. We show here that AKAP149 is a self-associating protein with RNA binding features. The KH domain of AKAP149 is sufficient for self-association in a RNA-dependent manner. The Tudor domain is not necessary for self-association, but it is required together with the KH domain for targeting to well-defined nuclear foci. These foci are spatially closely related to nucleolar subcompartments. We also show that the KH-Tudor-containing domain of AKAP149 binds RNA in vitro and in RNA coprecipitation experiments. AKAP149 emerges as a scaffolding protein involved in the integration of intracellular signals and possibly in RNA metabolism.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Dobramento de Proteína , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Ancoragem à Quinase A , Proteínas Adaptadoras de Transdução de Sinal/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína/genética , Transporte Proteico/fisiologia , Proteínas de Ligação a RNA/genética
13.
Symp Soc Exp Biol ; (56): 245-63, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15565886

RESUMO

The cell nucleus is a highly dynamic organelle whose function and structure during the cell cycle is tightly controlled. A number of signals triggered by external stimuli or intracellular clocks are relayed to the nucleus by protein kinases and phosphatases. Specificity of action of kinases and phosphatases can be achieved by their recruitment into multiprotein complexes targeted to discrete subcellular or subnuclear loci. One class of molecules targeting signalling units within single complexes are A-kinase anchoring proteins or AKAPs. AKAPs not only target enzymes to their substrate but may also regulate enzyme activity. This chapter highlights the role of nuclear AKAPs in relaying and modulating protein kinase and phosphatase signals to the nucleus or chromosomes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Núcleo Celular/fisiologia , Transdução de Sinais/fisiologia , Animais , Cálcio/fisiologia , Ciclo Celular/fisiologia , Cromatina/metabolismo , AMP Cíclico/fisiologia , Humanos , Camundongos , Membrana Nuclear/metabolismo
14.
J Cell Sci ; 116(Pt 11): 2237-46, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12697839

RESUMO

Reassembly of the nuclear envelope (NE) at the end of mitosis requires targeting of the B-type lamin protein phosphatase, PP1, to the envelope by A-kinase anchoring protein AKAP149. We show here that NE-associated AKAP149 is a novel PP1-specifying subunit involved in maintaining nuclear architecture through G1 phase. PP1 remains associated with NE-bound AKAP149 during G1 but is released from AKAP149 upon S phase entry, as AKAP149 becomes serine-phosphorylated. NE-associated AKAP149 inhibits PP1 activity towards glycogen phosphorylase but enhances PP1 phosphatase activity towards B-type lamins, indicating that AKAP149 is a B-type lamin specifying subunit of PP1. In vivo dissociation of PP1 from NE-bound AKAP149 in G1-phase nuclei triggers phosphorylation and depolymerization of A- and B-type lamins. The lamins solubilize intranuclearly without affecting the inner nuclear membrane or pore complex distribution. This correlates with the induction of a G1 arrest and, ultimately, apoptosis. We propose that AKAP149-regulated PP1 activity at the NE during G1 is required to maintain nuclear integrity and cell survival.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Fase G1/fisiologia , Proteínas de Membrana/metabolismo , Membrana Nuclear/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas de Ancoragem à Quinase A , Sobrevivência Celular/fisiologia , DNA/biossíntese , Células HeLa , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Fosforilação , Ligação Proteica/fisiologia , Solubilidade , Especificidade por Substrato
15.
EMBO Rep ; 3(4): 384-9, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11897658

RESUMO

We have developed a somatic cell-free system that remodels chromatin and activates gene expression in heterologous differentiated nuclei. Extracts of stimulated human T cells elicit chromatin binding of transcriptional activators of the interleukin-2 (IL-2) gene, anchoring and activity of a chromatin-remodeling complex and hyperacetylation of the IL-2 promoter in purified exogenous resting T-cell nuclei. The normally repressed IL-2 gene is transcribed in nuclei from quiescent human T cells and from various non-T-cell lines. This demonstrates that somatic cell extracts can be used to reprogram gene expression in differentiated nuclei. In vitro reprogramming may be useful for investigating regulation of gene expression and for producing replacement cells for the treatment of a wide variety of diseases.


Assuntos
Regulação da Expressão Gênica , Interleucina-2/genética , Linfócitos T/metabolismo , Acetilação , Acetiltransferases , Núcleo Celular/metabolismo , Sistema Livre de Células , Células Cultivadas , Cromatina/metabolismo , Células HeLa , Humanos , Ativação Linfocitária , Substâncias Macromoleculares , Regiões Promotoras Genéticas , Linfócitos T/imunologia , Fatores de Transcrição/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa