Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
CBE Life Sci Educ ; 17(3): es12, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30142049

RESUMO

The BioHealth Capital Region (Maryland, Virginia, and Washington, DC; BHCR) is flush with colleges and universities training students in science, technology, engineering, and mathematics disciplines and has one of the most highly educated workforces in the United States. However, current educational approaches and business recruitment tactics are not drawing sufficient talent to sustain the bioscience workforce pipeline. Surveys conducted by the Mid-Atlantic Biology Research and Career Network identified a disconnect between stakeholders who are key to educating, training, and hiring college and university graduates, resulting in several impediments to workforce development in the BHCR: 1) students are underinformed or unaware of bioscience opportunities before entering college and remain so at graduation; 2) students are not job ready at the time of graduation; 3) students are mentored to pursue education beyond what is needed and are therefore overqualified (by degree) for most of the available jobs in the region; 4) undergraduate programs generally lack any focus on workforce development; and 5) few industry-academic partnerships with undergraduate institutions exist in the region. The reality is that these issues are neither surprising nor restricted to the BHCR. Recommendations are presented to facilitate improvement in the preparation of graduates for today's bioscience industries throughout the United States.


Assuntos
Disciplinas das Ciências Biológicas/educação , Indústrias , Estudantes , Recursos Humanos , Escolha da Profissão , Aconselhamento , Humanos , Conhecimento , Percepção , Inquéritos e Questionários , Universidades
2.
BMC Mol Biol ; 16: 12, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-26063178

RESUMO

BACKGROUND: SIRT6, a member of the NAD(+)-dependent histone/protein deacetylase family, regulates genomic stability, metabolism, and lifespan. MYH glycosylase and APE1 are two base excision repair (BER) enzymes involved in mutation avoidance from oxidative DNA damage. Rad9-Rad1-Hus1 (9-1-1) checkpoint clamp promotes cell cycle checkpoint signaling and DNA repair. BER is coordinated with the checkpoint machinery and requires chromatin remodeling for efficient repair. SIRT6 is involved in DNA double-strand break repair and has been implicated in BER. Here we investigate the direct physical and functional interactions between SIRT6 and BER enzymes. RESULTS: We show that SIRT6 interacts with and stimulates MYH glycosylase and APE1. In addition, SIRT6 interacts with the 9-1-1 checkpoint clamp. These interactions are enhanced following oxidative stress. The interdomain connector of MYH is important for interactions with SIRT6, APE1, and 9-1-1. Mutagenesis studies indicate that SIRT6, APE1, and Hus1 bind overlapping but different sequence motifs on MYH. However, there is no competition of APE1, Hus1, or SIRT6 binding to MYH. Rather, one MYH partner enhances the association of the other two partners to MYH. Moreover, APE1 and Hus1 act together to stabilize the MYH/SIRT6 complex. Within human cells, MYH and SIRT6 are efficiently recruited to confined oxidative DNA damage sites within transcriptionally active chromatin, but not within repressive chromatin. In addition, Myh foci induced by oxidative stress and Sirt6 depletion are frequently localized on mouse telomeres. CONCLUSIONS: Although SIRT6, APE1, and 9-1-1 bind to the interdomain connector of MYH, they do not compete for MYH association. Our findings indicate that SIRT6 forms a complex with MYH, APE1, and 9-1-1 to maintain genomic and telomeric integrity in mammalian cells.


Assuntos
Pontos de Checagem do Ciclo Celular , Reparo do DNA , DNA/metabolismo , Sirtuínas/metabolismo , Motivos de Aminoácidos , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , DNA Glicosilases/genética , DNA Glicosilases/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Exonucleases/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Sirtuínas/genética , Telômero/metabolismo
3.
DNA Repair (Amst) ; 31: 80-90, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26021743

RESUMO

Cell cycle checkpoints provide surveillance mechanisms to activate the DNA damage response, thus preserving genomic integrity. The heterotrimeric Rad9-Rad1-Hus1 (9-1-1) clamp is a DNA damage response sensor and can be loaded onto DNA. 9-1-1 is involved in base excision repair (BER) by interacting with nearly every enzyme in BER. Here, we show that individual 9-1-1 components play distinct roles in BER directed by MYH DNA glycosylase. Analyses of Hus1 deletion mutants revealed that the interdomain connecting loop (residues 134-155) is a key determinant of MYH binding. Both the N-(residues 1-146) and C-terminal (residues 147-280) halves of Hus1, which share structural similarity, can interact with and stimulate MYH. The Hus1(K136A) mutant retains physical interaction with MYH but cannot stimulate MYH glycosylase activity. The N-terminal domain, but not the C-terminal half of Hus1 can also bind DNA with moderate affinity. Intact Rad9 expressed in bacteria binds to and stimulates MYH weakly. However, Rad9(1-266) (C-terminal truncated Rad9) can stimulate MYH activity and bind DNA with high affinity, close to that displayed by heterotrimeric 9(1-266)-1-1 complexes. Conversely, Rad1 has minimal roles in stimulating MYH activity or binding to DNA. Finally, we show that preferential recruitment of 9(1-266)-1-1 to 5'-recessed DNA substrates is an intrinsic property of this complex and is dependent on complex formation. Together, our findings provide a mechanistic rationale for unique contributions by individual 9-1-1 subunits to MYH-directed BER based on subunit asymmetry in protein-protein interactions and DNA binding events.


Assuntos
Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/genética , DNA Glicosilases/genética , Exonucleases/genética , Animais , Sítios de Ligação , Proteínas de Ciclo Celular/metabolismo , Clonagem Molecular , DNA Glicosilases/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Exonucleases/metabolismo , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Camundongos , Conformação Proteica
4.
Am J Cancer Res ; 4(3): 211-21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24959376

RESUMO

SIRT1, a member of the NAD(+)-dependent histone/protein deacetylase family, is involved in chromatin remodeling, DNA repair, and stress response and is a potential drug target. 5-fluorouracil (FU) and the SN1-type DNA methylating agent temozolomide (TMZ) are anticancer agents. In this study, we demonstrate that sirt1 knockout mouse embryonic fibroblast cells are more sensitive to FU and DNA methylating agents than normal cells. Based on these findings, the chemotherapy efficacy of SIRT1 inhibitors in combination with FU or TMZ were tested with human breast cancer cells. We found that treatments combining SIRT1 inhibitors with FU or TMZ show synergistic reduction of cell viability and colony formation of breast cancer cells. Thus, inhibition of SIRT1 activity provides a novel anticancer strategy.

5.
Biochem J ; 456(1): 89-98, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23952905

RESUMO

TDG (thymine DNA glycosylase) is an essential multifunctional enzyme involved in DNA base excision repair, DNA demethylation and transcription regulation. TDG is the predominant enzyme that removes thymine from T/G mispair, which arises due to deamination of 5-methyl-cytosine at the CpG dinucleotide, thereby preventing C to T mutations. SIRT1 is a member of class III NAD+-dependent histone/protein deacetylases. In the present study, we demonstrate that SIRT1 interacts with residues 67-110 of hTDG (human TDG). In addition, SIRT1 enhances TDG glycosylase activity and deacetylates acetylated TDG. TDG acetylation weakens its interaction with SIRT1. Although acetylated TDG has reduced glycosylase activity towards T/G, 5-formylcytosine/G and 5-carboxylcytosine/G, it has a stronger activity towards a 5-fluorouracil/G substrate as compared with unmodified TDG. SIRT1 weakly stimulates acetylated hTDG activity towards T/G, 5-formylcytosine/G and 5-carboxylcytosine/G as compared with control hTDG. Sirt1-knockout mouse embryonic fibroblast cells have higher levels of TDG expression and acetylation. The physical and functional interactions between SIRT1 and TDG may mediate DNA repair, gene expression and FU (5-fluorouracil)-mediated cytotoxicity.


Assuntos
Sirtuína 1/metabolismo , Timina DNA Glicosilase/metabolismo , Acetilação , Animais , Antineoplásicos/química , Reparo do DNA , Fibroblastos/metabolismo , Fluoruracila/química , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Sirtuína 1/química , Sirtuína 1/genética , Especificidade por Substrato , Timina DNA Glicosilase/química
6.
J Mol Biol ; 403(3): 351-70, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20816984

RESUMO

The DNA glycosylase MutY homologue (MYH or MUTYH) removes adenines misincorporated opposite 8-oxoguanine as part of the base excision repair pathway. Importantly, defects in human MYH (hMYH) activity cause the inherited colorectal cancer syndrome MYH-associated polyposis. A key feature of MYH activity is its coordination with cell cycle checkpoint via interaction with the Rad9-Rad1-Hus1 (9-1-1) complex. The 9-1-1 complex facilitates cell cycle checkpoint activity and coordinates this activity with ongoing DNA repair. The interdomain connector (IDC, residues 295-350) between the catalytic domain and the 8-oxoguanine recognition domain of hMYH is a critical element that maintains interactions with the 9-1-1 complex. We report the first crystal structure of a eukaryotic MutY protein, a fragment of hMYH (residues 65-350) that consists of the catalytic domain and the IDC. Our structure reveals that the IDC adopts a stabilized conformation projecting away from the catalytic domain to form a docking scaffold for 9-1-1. We further examined the role of the IDC using Schizosaccharomyces pombe MYH as model system. In vitro studies of S. pombe MYH identified residues I261 and E262 of the IDC (equivalent to V315 and E316 of the hMYH IDC) as critical for maintaining the MYH/9-1-1 interaction. We determined that the eukaryotic IDC is also required for DNA damage selection and robust enzymatic activity. Our studies also provide the first evidence that disruption of the MYH/9-1-1 interaction diminishes the repair of oxidative DNA damage in vivo. Thus, preserving the MYH/9-1-1 interaction contributes significantly to minimizing the mutagenic potential of oxidative DNA damage.


Assuntos
Proteínas de Ciclo Celular/metabolismo , DNA Glicosilases/química , DNA Glicosilases/metabolismo , Exonucleases/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas de Ciclo Celular/genética , DNA/genética , DNA/metabolismo , Dano ao DNA , DNA Glicosilases/genética , Reparo do DNA , Exonucleases/genética , Humanos , Peróxido de Hidrogênio/metabolismo , Mutação/genética , Ligação Proteica , Schizosaccharomyces/genética , Schizosaccharomyces/crescimento & desenvolvimento , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética
7.
DNA Repair (Amst) ; 9(5): 478-87, 2010 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-20188637

RESUMO

In eukaryotic cells, the cell cycle checkpoint proteins Rad9, Rad1, and Hus1 form the 9-1-1 complex which is structurally similar to the proliferating cell nuclear antigen (PCNA) sliding clamp. hMSH2/hMSH6 (hMutS alpha) and hMSH2/hMSH3 (hMutS beta) are the mismatch recognition factors of the mismatch repair pathway. hMutS alpha has been shown to physically and functionally interact with PCNA. Moreover, DNA methylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) treatment induces the G2/M cell cycle arrest that is dependent on the presence of hMutS alpha and hMutL alpha. In this study, we show that each subunit of the human 9-1-1 complex physically interacts with hMSH2, hMSH3, and hMSH6. The 9-1-1 complex from both humans and Schizosaccharomyces pombe can stimulate hMutS alpha binding with G/T-containing DNA. Rad9, Rad1, and Hus1 individual subunits can also stimulate the DNA binding activity of hMutS alpha. Human Rad9 and hMSH6 colocalize to nuclear foci of HeLa cells after exposure to MNNG. However, Rad9 does not form foci in MSH6 defective cells following MNNG treatment. In Rad9 knockdown untreated cells, the majority of the MSH6 is in cytoplasm. Following MNNG treatment, Rad9 knockdown cells has abnormal nuclear morphology and MSH6 is distributed around nuclear envelop. Our findings suggest that the 9-1-1 complex is a component of the mismatch repair involved in MNNG-induced damage response.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Reparo de Erro de Pareamento de DNA , Proteínas de Ligação a DNA/metabolismo , Exonucleases/metabolismo , Pareamento Incorreto de Bases , Sequência de Bases , Núcleo Celular/metabolismo , Metilação de DNA/efeitos dos fármacos , Células HeLa , Humanos , Metilnitronitrosoguanidina/farmacologia , Dados de Sequência Molecular , Subunidades Proteicas/metabolismo , Transporte Proteico
8.
BMC Biochem ; 10: 19, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19523222

RESUMO

BACKGROUND: Escherichia coli MutY (EcMutY) reduces mutagenesis by removing adenines paired with guanines or 7,8-dihydro-8-oxo-guanines (8-oxoG). V45 and Q182 of EcMutY are considered to be the key determinants of adenine specificity. Both residues are spatially close to each other in the active site and are conserved in MutY family proteins but not in Methanobacterium thermoautotrophicum Mig.MthI T/G mismatch DNA glycosylase (A50 and L187 at the corresponding respective positions). RESULTS: Targeted mutagenesis study was performed to determine the substrate specificities of V45A, Q182L, and V45A/Q182L double mutant of EcMutY. All three mutants had significantly lower binding and glycosylase activities for A/G and A/8-oxoG mismatches than the wild-type enzyme. The double mutant exhibited an additive reduction in binding to both the A/G and A/GO in comparison to the single mutants. These mutants were also tested for binding and glycosylase activities with T/G- or T/8-oxoG-containing DNA. Both V45A and Q182L mutants had substantially increased affinities towards T/G, however, they did not exhibit any T/G or T/8-oxoG glycosylase activity. Surprisingly, the V45A/Q182L double mutant had similar binding affinities to T/G as the wild-type EcMutY. V45A, Q182L, and V45A/Q182L EcMutY mutants could not reduce the G:C to T:A mutation frequency of a mutY mutant. Expression of the V45A mutant protein caused a dominant negative phenotype with an increased G:C to A:T mutation frequency. CONCLUSION: The substrate specificities are altered in V45A, Q182L, and V45A/Q182L EcMutY mutants. V45A and Q182L mutants had reduced binding and glycosylase activities for A/G and A/8-oxoG mismatches and increased affinities towards T/G mismatch. However, in contrast to a previous report that Mig.MthI thymine DNA glycosylase can be converted to a MutY-like adenine glycosylase by replacing two residues (A50V and L187Q), both V45A and Q182L EcMutY mutants did not exhibit any T/G or T/8-oxoG glycosylase activity. The dominant negative phenotype of V45A EcMutY mutant protein is probably caused by its increased binding affinity to T/G mismatch and thus inhibiting other repair pathways.


Assuntos
DNA Glicosilases/química , DNA Glicosilases/metabolismo , Escherichia coli/enzimologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , DNA Glicosilases/genética , Reparo de Erro de Pareamento de DNA , Escherichia coli/química , Escherichia coli/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Ligação Proteica , Alinhamento de Sequência , Especificidade por Substrato
9.
Nucleic Acids Res ; 35(18): 6207-18, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17855402

RESUMO

Human (h) DNA repair enzyme thymine DNA glycosylase (hTDG) is a key DNA glycosylase in the base excision repair (BER) pathway that repairs deaminated cytosines and 5-methyl-cytosines. The cell cycle checkpoint protein Rad9-Rad1-Hus1 (the 9-1-1 complex) is the surveillance machinery involved in the preservation of genome stability. In this study, we show that hTDG interacts with hRad9, hRad1 and hHus1 as individual proteins and as a complex. The hHus1 interacting domain is mapped to residues 67-110 of hTDG, and Val74 of hTDG plays an important role in the TDG-Hus1 interaction. In contrast to the core domain of hTDG (residues 110-308), hTDG(67-308) removes U and T from U/G and T/G mispairs, respectively, with similar rates as native hTDG. Human TDG activity is significantly stimulated by hHus1, hRad1, hRad9 separately, and by the 9-1-1 complex. Interestingly, the interaction between hRad9 and hTDG, as detected by co-immunoprecipitation (Co-IP), is enhanced following N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) treatment. A significant fraction of the hTDG nuclear foci co-localize with hRad9 foci in cells treated with methylating agents. Thus, the 9-1-1 complex at the lesion sites serves as both a damage sensor to activate checkpoint control and a component of the BER.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Reparo do DNA , Exonucleases/metabolismo , Timina DNA Glicosilase/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Dano ao DNA , Ativação Enzimática , Células HeLa , Humanos , Dados de Sequência Molecular , Timina DNA Glicosilase/análise , Timina DNA Glicosilase/química
10.
Acta Crystallogr D Biol Crystallogr ; 62(Pt 3): 246-52, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16510971

RESUMO

The malarial parasite continues to be one of the leading causes of death in many developing countries. With the development of resistance to the currently available treatments, the discovery of new therapeutics is imperative. Currently, the plasmepsin enzymes found in the food vacuole of the parasite are a chief target for drug development. Allophenylnorstatine-based compounds originally designed to inhibit HIV-1 protease have shown efficacy against all four plasmepsin enzymes found in the food vacuole of Plasmodium falciparum. In this study, the first crystal structure of P. malariae plasmepsin 4 (PmPM4) bound to the allophenylnorstatine-based compound KNI-764 is described at 3.3 Angstroms resolution. The PmPM4-inhibitor complex crystallized in the orthorhombic space group P2(1)2(1)2, with unit-cell parameters a = 95.9, b = 112.6, c = 90.4 Angstroms, with two molecules in the asymmetric unit related by a non-crystallographic symmetry operator. The structure was refined to a final R factor of 24.7%. The complex showed the inhibitor in an unexpected binding orientation with allophenylnorstatine occupying the S1' pocket. The P2 group was found outside the S2 pocket, wedged between the flap and a juxtaposed loop. Inhibition analysis of PmPM4 also suggests the potential for allophenylnorstatine-based compounds to be effective against all species of malaria infecting humans and for the future development of a broad-based inhibitor.


Assuntos
Antimaláricos/metabolismo , Ácido Aspártico Endopeptidases/química , Fenilbutiratos/metabolismo , Plasmodium malariae/química , Inibidores de Proteases/metabolismo , Animais , Antimaláricos/química , Ácido Aspártico Endopeptidases/isolamento & purificação , Cristalização , Cristalografia por Raios X , Cinética , Modelos Moleculares , Conformação Molecular , Fenilbutiratos/química , Inibidores de Proteases/química , Ligação Proteica
11.
Biochemistry ; 44(6): 1768-79, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15697202

RESUMO

Two targeted chromogenic octapeptide combinatorial libraries, comprised of 38 pools each containing 361 different peptides, were used to analyze the enzyme/substrate interactions of five plasmepsins. The first library (P1 library) was based on a good synthetic aspartic peptidase substrate [Westling, J., Cipullo, P., Hung, S. H., Saft, H., Dame, J. B., and Dunn, B. M. (1999) Protein Sci. 8, 2001-2009; Scarborough, P. E., and Dunn, B. M. (1994) Protein Eng. 7, 495-502] and had the sequence Lys-Pro-(Xaa)-Glu-P1*Nph-(Xaa)-Leu. The second library (P1' library) incorporated results with the plasmepsins from the first library and had the sequence Lys-Pro-Ile-(Xaa)-Nph*P1'-Gln-(Xaa). In both cases, P1 and P1' were fixed residues for a given peptide pool, where Nph was a para-nitrophenylalanine chromogenic reporter and Xaa was a mixture of 19 different amino acids. Kinetic assays monitoring the rates of cleavage of these libraries revealed the optimal P1 and P1' residues for the five plasmepsins as hydrophobic substitutions. Extended specificity preferences were obtained utilizing liquid chromatography-mass spectrometry (LC-MS) to analyze the cleavage products produced by enzyme-catalyzed digestion of the best pools of each peptide library. LC-MS analysis of the P1-Phe and P1'-Phe pools revealed the favored amino acids at the P3, P2, P2', and P3' positions. These analyses have provided new insights on the binding preferences of malarial digestive enzymes that were used to design specific methyleneamino peptidomimetic inhibitors of the plasmepsins. Some of these compounds were potent inhibitors of the five plasmepsins, and their possible binding modes were analyzed by computational methods.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Compostos Cromogênicos/metabolismo , Técnicas de Química Combinatória/métodos , Malária/enzimologia , Biblioteca de Peptídeos , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/síntese química , Sítios de Ligação , Compostos Cromogênicos/química , Biologia Computacional/métodos , Simulação por Computador , Hidrólise , Modelos Moleculares , Plasmodium falciparum/enzimologia , Plasmodium malariae/enzimologia , Plasmodium ovale/enzimologia , Plasmodium vivax/enzimologia , Inibidores de Proteases/síntese química , Inibidores de Proteases/farmacologia , Proteínas de Protozoários , Especificidade por Substrato
12.
Artigo em Inglês | MEDLINE | ID: mdl-16511002

RESUMO

Plasmepsin 4 from the malarial parasite Plasmodium malariae (PmPM4) is a member of the plasmepsins (Plasmodium pepsins), a subfamily of the pepsin-like aspartic proteases whose ortholog in the malarial parasite P. falciparum is involved in hemoglobin digestion in its digestive vacuole. Crystals of PmPM4 in complex with the small-molecule inhibitor AG1776 have been grown from a precipitant of 15% PEG 4000 and 200 mM ammonium sulfate in 100 mM sodium acetate pH 4.5. X-ray diffraction data were collected on a Rigaku rotating-anode generator from a single crystal under cryoconditions, with a maximal useful diffraction pattern to 3.3 A resolution. The crystals are shown to be orthorhombic and have been assigned to space group P2(1)2(1)2, with unit-cell parameters a = 95.88, b = 112.58, c = 90.40 A and a scaling Rsym of 0.104 for 14,334 unique reflections. Packing consideration and self-rotation function results indicate that there are two molecules per asymmetric unit. It is expected that in the near future the structure of PmPM4 will be obtained using molecular-replacement methods, obtaining phases from previously determined plasmepsin structures. Elucidation of the structure of PmPM4 in complex with inhibitors may be paramount to producing new antimalarial therapeutic agents.


Assuntos
Ácido Aspártico Endopeptidases/química , Plasmodium malariae/enzimologia , Sequência de Aminoácidos , Animais , Ácido Aspártico Endopeptidases/isolamento & purificação , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/isolamento & purificação , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA