Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nucleic Acids Res ; 45(3): 1442-1454, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28180316

RESUMO

RIG-I is an innate immune receptor that detects and responds to infection by deadly RNA viruses such as influenza, and Hepatitis C. In the cytoplasm, RIG-I is faced with a difficult challenge: it must sensitively detect viral RNA while ignoring the abundance of host RNA. It has been suggested that RIG-I has a 'proof-reading' mechanism for rejecting host RNA targets, and that disruptions of this selectivity filter give rise to autoimmune diseases. Here, we directly monitor RNA proof-reading by RIG-I and we show that it is controlled by a set of conserved amino acids that couple RNA and ATP binding to the protein (Motif III). Mutations of this motif directly modulate proof-reading by eliminating or enhancing selectivity for viral RNA, with major implications for autoimmune disease and cancer. More broadly, the results provide a physical explanation for the ATP-gated behavior of SF2 RNA helicases and receptor proteins.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteína DEAD-box 58/metabolismo , RNA/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Substituição de Aminoácidos , Autoimunidade , Sítios de Ligação/genética , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Células HEK293 , Humanos , Imunidade Inata , Modelos Moleculares , Mutagênese Sítio-Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Domínios e Motivos de Interação entre Proteínas , RNA/química , RNA/genética , Vírus de RNA/genética , Vírus de RNA/imunologia , Vírus de RNA/patogenicidade , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Receptores Imunológicos , Receptores de Reconhecimento de Padrão/química , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais
2.
Proc Natl Acad Sci U S A ; 112(3): 827-32, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25561542

RESUMO

Most isolates of human rhinovirus, the common cold virus, replicate more robustly at the cool temperatures found in the nasal cavity (33-35 °C) than at core body temperature (37 °C). To gain insight into the mechanism of temperature-dependent growth, we compared the transcriptional response of primary mouse airway epithelial cells infected with rhinovirus at 33 °C vs. 37 °C. Mouse airway cells infected with mouse-adapted rhinovirus 1B exhibited a striking enrichment in expression of antiviral defense response genes at 37 °C relative to 33 °C, which correlated with significantly higher expression levels of type I and type III IFN genes and IFN-stimulated genes (ISGs) at 37 °C. Temperature-dependent IFN induction in response to rhinovirus was dependent on the MAVS protein, a key signaling adaptor of the RIG-I-like receptors (RLRs). Stimulation of primary airway cells with the synthetic RLR ligand poly I:C led to greater IFN induction at 37 °C relative to 33 °C at early time points poststimulation and to a sustained increase in the induction of ISGs at 37 °C relative to 33 °C. Recombinant type I IFN also stimulated more robust induction of ISGs at 37 °C than at 33 °C. Genetic deficiency of MAVS or the type I IFN receptor in infected airway cells permitted higher levels of viral replication, particularly at 37 °C, and partially rescued the temperature-dependent growth phenotype. These findings demonstrate that in mouse airway cells, rhinovirus replicates preferentially at nasal cavity temperature due, in part, to a less efficient antiviral defense response of infected cells at cool temperature.


Assuntos
Imunidade Inata , Rhinovirus/imunologia , Traqueia/virologia , Replicação Viral , Animais , Camundongos , Dados de Sequência Molecular , Rhinovirus/fisiologia , Temperatura
3.
Nucleic Acids Res ; 42(6): 3919-30, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24435798

RESUMO

Dicer is a specialized nuclease that produces RNA molecules of specific lengths for use in gene silencing pathways. Dicer relies on the correct measurement of RNA target duplexes to generate products of specific lengths. It is thought that Dicer uses its multidomain architecture to calibrate RNA product length. However, this measurement model is derived from structural information from a protozoan Dicer, and does not account for the helicase domain present in higher organisms. The Caenorhabditis elegans Dicer-related helicase 3 (DRH-3) is an ortholog of the Dicer and RIG-I family of double-strand RNA activated ATPases essential for secondary siRNA production. We find that DRH-3 specifies 22 bp RNAs by dimerization of the helicase domain, a process mediated by ATPase activity and the N-terminal domain. This mechanism for RNA length discrimination by a Dicer family protein suggests an alternative model for RNA length measurement by Dicer, with implications for recognition of siRNA and miRNA targets.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , RNA Helicases DEAD-box/metabolismo , RNA Helicases/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Caenorhabditis elegans/química , RNA Helicases DEAD-box/química , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , RNA Helicases/química , RNA de Cadeia Dupla/química , RNA Interferente Pequeno/química
4.
J Biol Chem ; 289(22): 15810-9, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24753249

RESUMO

Thymine DNA glycosylase (TDG) initiates the repair of G·T mismatches that arise by deamination of 5-methylcytosine (mC), and it excises 5-formylcytosine and 5-carboxylcytosine, oxidized forms of mC. TDG functions in active DNA demethylation and is essential for embryonic development. TDG forms a tight enzyme-product complex with abasic DNA, which severely impedes enzymatic turnover. Modification of TDG by small ubiquitin-like modifier (SUMO) proteins weakens its binding to abasic DNA. It was proposed that sumoylation of product-bound TDG regulates product release, with SUMO conjugation and deconjugation needed for each catalytic cycle, but this model remains unsubstantiated. We examined the efficiency and specificity of TDG sumoylation using in vitro assays with purified E1 and E2 enzymes, finding that TDG is modified efficiently by SUMO-1 and SUMO-2. Remarkably, we observed similar modification rates for free TDG and TDG bound to abasic or undamaged DNA. To examine the conjugation step directly, we determined modification rates (kobs) using preformed E2∼SUMO-1 thioester. The hyperbolic dependence of kobs on TDG concentration gives kmax = 1.6 min(-1) and K1/2 = 0.55 µM, suggesting that E2∼SUMO-1 has higher affinity for TDG than for the SUMO targets RanGAP1 and p53 (peptide). Whereas sumoylation substantially weakens TDG binding to DNA, TDG∼SUMO-1 still binds relatively tightly to AP-DNA (Kd ∼50 nM). Although E2∼SUMO-1 exhibits no specificity for product-bound TDG, the relatively high conjugation efficiency raises the possibility that E2-mediated sumoylation could stimulate product release in vivo. This and other implications for the biological role and mechanism of TDG sumoylation are discussed.


Assuntos
Reparo do DNA/fisiologia , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Timina DNA Glicosilase/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Metilação de DNA/fisiologia , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Glicosilação , Células HeLa , Humanos , Processamento de Proteína Pós-Traducional/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteína SUMO-1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Especificidade por Substrato , Sumoilação/fisiologia , Timina DNA Glicosilase/química , Timina DNA Glicosilase/genética , Enzimas de Conjugação de Ubiquitina/genética
5.
bioRxiv ; 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38915482

RESUMO

Lysine Specific Demethylase 1 (KDM1A / LSD1) regulates mitochondrial respiration and stabilizes HIF-1A (hypoxia-inducible factor 1A). HIF-1A modulates reactive oxygen species (ROS) levels by increasing cellular glucose uptake, glycolysis, and endogenous antioxidants. The role of KDM1A in cellular ROS response has not previously been described. We determined the role of KDM1A in regulating the ROS response and the utility of KDM1A inhibitors in combination with ROS-inducing cancer therapies. Our results show that KDM1A inhibition sensitized cells to oxidative stress and increased total cellular ROS, which was mitigated by treatment with the antioxidant N-acetyl cysteine. KDM1A inhibition decreased basal mitochondrial respiration and impaired induction of HIF-1A after ROS exposure. Overexpression of HIF-1A salvaged cells from KDM1A inhibition enhanced sensitivity to ROS. Thus we found that increased sensitivity of ROS after KDM1A inhibition was mediated by HIF-1A and depletion of endogenous glutathione. We also show that KDM1A-specific inhibitor bizine synergized with antioxidant-depleting therapies, buthionine sulfoximine, and auranofin in rhabdomyosarcoma cell lines (Rh28 and Rh30). In this study, we describe a novel role for KDM1A in regulating HIF-1A functions under oxidative stress and found that dual targeting of KDM1A and antioxidant systems may serve as an effective combination anticancer strategy.

6.
Nucleic Acids Res ; 39(6): 2319-29, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21097883

RESUMO

Deamination of 5-methylcytosine to thymine creates mutagenic G · T mispairs, contributing to cancer and genetic disease. Thymine DNA glycosylase (TDG) removes thymine from these G · T lesions, and follow-on base excision repair yields a G · C pair. A previous crystal structure revealed TDG (catalytic domain) bound to abasic DNA product in a 2:1 complex, one subunit at the abasic site and the other bound to undamaged DNA. Biochemical studies showed TDG can bind abasic DNA with 1:1 or 2:1 stoichiometry, but the dissociation constants were unknown, as was the stoichiometry and affinity for binding substrates and undamaged DNA. We showed that 2:1 binding is dispensable for G · U activity, but its role in G · T repair was unknown. Using equilibrium binding anisotropy experiments, we show that a single TDG subunit binds very tightly to G · U mispairs and abasic (G · AP) sites, and somewhat less tightly G · T mispairs. Kinetics experiments show 1:1 binding provides full G · T activity. TDG binds undamaged CpG sites with remarkable affinity, modestly weaker than G · T mispairs, and exhibits substantial affinity for nonspecific DNA. While 2:1 binding is observed for large excess TDG concentrations, our findings indicate that a single TDG subunit is fully capable of locating and processing G · U or G · T lesions.


Assuntos
Pareamento Incorreto de Bases , DNA/metabolismo , Timina DNA Glicosilase/metabolismo , Ilhas de CpG , DNA/química , Dano ao DNA , Reparo do DNA , Cinética , Ligação Proteica , Timina DNA Glicosilase/química
7.
Cancers (Basel) ; 15(5)2023 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-36900402

RESUMO

Mutations found in skin are acquired in specific patterns, clustering around mutation-prone genomic locations. The most mutation-prone genomic areas, mutation hotspots, first induce the growth of small cell clones in healthy skin. Mutations accumulate over time, and clones with driver mutations may give rise to skin cancer. Early mutation accumulation is a crucial first step in photocarcinogenesis. Therefore, a sufficient understanding of the process may help predict disease onset and identify avenues for skin cancer prevention. Early epidermal mutation profiles are typically established using high-depth targeted next-generation sequencing. However, there is currently a lack of tools for designing custom panels to capture mutation-enriched genomic regions efficiently. To address this issue, we created a computational algorithm that implements a pseudo-exhaustive approach to identify the best genomic areas to target. We benchmarked the current algorithm in three independent mutation datasets of human epidermal samples. Compared to the sequencing panel designs originally used in these publications, the mutation capture efficacy (number of mutations/base pairs sequenced) of our designed panel improved 9.6-12.1-fold. Mutation burden in the chronically sun-exposed and intermittently sun-exposed normal epidermis was measured within genomic regions identified by hotSPOT based on cutaneous squamous cell carcinoma (cSCC) mutation patterns. We found a significant increase in mutation capture efficacy and mutation burden in cSCC hotspots in chronically sun-exposed vs. intermittently sun-exposed epidermis (p < 0.0001). Our results show that our hotSPOT web application provides a publicly available resource for researchers to design custom panels, enabling efficient detection of somatic mutations in clinically normal tissues and other similar targeted sequencing studies. Moreover, hotSPOT also enables the comparison of mutation burden between normal tissues and cancer.

8.
Sci Adv ; 7(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33523857

RESUMO

In ultraviolet (UV) radiation-exposed skin, mutations fuel clonal cell growth. The relationship between UV exposure and the accumulation of clonal mutations (CMs) and the correlation between CMs and skin cancer risk are largely unexplored. We characterized 450 individual-matched sun-exposed (SE) and non-SE (NE) normal human skin samples. The number and relative contribution of CMs were significantly different between SE and NE areas. Furthermore, we identified hotspots in TP53, NOTCH1, and GRM3 where mutations were significantly associated with UV exposure. In the normal skin from patients with cutaneous squamous cell carcinoma, we found that the cancer burden was associated with the UV-induced mutations, with the difference mostly conferred by the low-frequency CMs. These findings provide previously unknown information on UV's carcinogenic effect and pave the road for future development of quantitative assessment of subclinical UV damage and skin cancer risk.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Cutâneas , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Humanos , Mutação , Pele/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Raios Ultravioleta/efeitos adversos
9.
Photodiagnosis Photodyn Ther ; 30: 101743, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32198019

RESUMO

Visual examination plays a central role in the diagnosis of skin diseases. Many dermatologists use magnification, or dermoscopy, to improve diagnostic certainty when assessing the skin under visible light. In addition to magnification, other technological advances have been made over the last century to improve our visual assessment of the skin. Examination of skin under ultraviolet (UV) radiation, with Wood's light, gained traction for its utility in assessing superficial cutaneous infections and pigmentary changes. During Wood's light examination, UV light is directed at the skin and fluorescence is detected by our eyes. The variable fluorescent characteristics of endogenous and exogenous cutaneous chromophores help us better diagnose skin disease. UV fluorescent photography is based on the same concept as the Wood's light, but also allows image analysis and documentation of the captured image. In addition to UV-induced fluorescence, the differential reflection and absorption of UV light captured in the UV spectral range can also provide a new contrast for diagnosing skin diseases during UV reflectance photography. This review discusses the most widely used UV imaging techniques and provides an overview of the role of UV imaging in dermatology.


Assuntos
Dermatologia , Fotoquimioterapia , Dermatopatias , Humanos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes , Dermatopatias/diagnóstico por imagem , Raios Ultravioleta
10.
Chem Biol ; 15(3): 203-4, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18355716

RESUMO

During reverse transcription, an RNA polypurine tract (PPT) resists digestion by reverse transcriptase (RT) and primes plus-strand DNA synthesis. In this issue of Chemistry & Biology, Yi-Brunozzi et al. (2008) report structural studies of PPTs, illuminating how they are recognized by RT.


Assuntos
DNA Viral/química , Nucleotídeos de Purina/química , RNA Viral/química , Composição de Bases , Sequência de Bases , Sítios de Ligação , DNA Viral/genética , DNA Viral/metabolismo , Transcriptase Reversa do HIV/metabolismo , Nucleotídeos de Purina/genética , Nucleotídeos de Purina/metabolismo , RNA Viral/genética , RNA Viral/metabolismo
11.
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
12.
Sci Adv ; 4(2): e1701854, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29492454

RESUMO

We have developed highly potent synthetic activators of the vertebrate immune system that specifically target the RIG-I receptor. When introduced into mice, a family of short, triphosphorylated stem-loop RNAs (SLRs) induces a potent interferon response and the activation of specific genes essential for antiviral defense. Using RNA sequencing, we provide the first in vivo genome-wide view of the expression networks that are initiated upon RIG-I activation. We observe that SLRs specifically induce type I interferons, subsets of interferon-stimulated genes (ISGs), and cellular remodeling factors. By contrast, polyinosinic:polycytidylic acid [poly(I:C)], which binds and activates multiple RNA sensors, induces type III interferons and several unique ISGs. The short length (10 to 14 base pairs) and robust function of SLRs in mice demonstrate that RIG-I forms active signaling complexes without oligomerizing on RNA. These findings demonstrate that SLRs are potent therapeutic and investigative tools for targeted modulation of the innate immune system.


Assuntos
Proteína DEAD-box 58/metabolismo , RNA/metabolismo , Células A549 , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Interferons/farmacologia , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Conformação de Ácido Nucleico , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , RNA/química , Receptores Imunológicos
13.
Elife ; 42015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26371557

RESUMO

Retinoic acid-inducible gene I (RIG-I) initiates a rapid innate immune response upon detection and binding to viral ribonucleic acid (RNA). This signal activation occurs only when pathogenic RNA is identified, despite the ability of RIG-I to bind endogenous RNA while surveying the cytoplasm. Here we show that ATP binding and hydrolysis by RIG-I play a key role in the identification of viral targets and the activation of signaling. Using biochemical and cell-based assays together with mutagenesis, we show that ATP binding, and not hydrolysis, is required for RIG-I signaling on viral RNA. However, we show that ATP hydrolysis does provide an important function by recycling RIG-I and promoting its dissociation from non-pathogenic RNA. This activity provides a valuable proof-reading mechanism that enhances specificity and prevents an antiviral response upon encounter with host RNA molecules.


Assuntos
Trifosfato de Adenosina/metabolismo , RNA Helicases DEAD-box/metabolismo , Imunidade Inata , RNA Viral/metabolismo , Transdução de Sinais , Linhagem Celular , Proteína DEAD-box 58 , Humanos , Hidrólise , Ligação Proteica , Receptores Imunológicos
14.
Curr Opin Microbiol ; 20: 76-81, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24912143

RESUMO

RIG-I-like receptors (RLRs) utilize a specialized, multi-domain architecture to detect and respond to invasion by a diverse set of viruses. Structural similarities among these receptors provide a general mechanism for double strand RNA recognition and signal transduction. However, each RLR has developed unique strategies for sensing the specific molecular determinants on subgroups of viral RNAs. As a means to circumvent the antiviral response, viruses escape RLR detection by degrading, or sequestering or modifying their RNA. Patterns of variation in RLR sequence reveal a continuous evolution of the protein domains that contribute to RNA recognition and signaling.


Assuntos
RNA Helicases DEAD-box/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Receptores Imunológicos/metabolismo , Animais , RNA Helicases DEAD-box/imunologia , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , RNA de Cadeia Dupla/imunologia , RNA Viral/imunologia , Receptores Imunológicos/imunologia
15.
J Biol Chem ; 283(47): 32680-90, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18805789

RESUMO

DNA glycosylases initiate base excision repair by removing damaged or mismatched bases, producing apurinic/apyrimidinic (AP) DNA. For many glycosylases, the AP-DNA remains tightly bound, impeding enzymatic turnover. A prominent example is thymine DNA glycosylase (TDG), which removes T from G.T mispairs and recognizes other lesions, with specificity for damage at CpG dinucleotides. TDG turnover is very slow; its activity appears to reach a plateau as the [product]/[enzyme] ratio approaches unity. The follow-on base excision repair enzyme, AP endonuclease 1 (APE1), stimulates the turnover of TDG and other glycosylases, involving a mechanism that remains largely unknown. We examined the catalytic activity of human TDG (hTDG), alone and with human APE1 (hAPE1), using pre-steady-state kinetics and a coupled-enzyme (hTDG-hAPE1) fluorescence assay. hTDG turnover is exceedingly slow for G.T (k(cat)=0.00034 min(-1)) and G.U (k(cat)=0.005 min(-1)) substrates, much slower than k(max) from single turnover experiments, confirming that AP-DNA release is rate-limiting. We find unexpectedly large differences in k(cat) for G.T, G.U, and G.FU substrates, indicating the excised base remains trapped in the product complex by AP-DNA. hAPE1 increases hTDG turnover by 42- and 26-fold for G.T and G.U substrates, the first quantitative measure of the effect of hAPE1. hAPE1 stimulates hTDG by disrupting the product complex rather than merely depleting (endonucleolytically) the AP-DNA. The enhancement is greater for hTDG catalytic core (residues 111-308 of 410), indicating the N- and C-terminal domains are dispensable for stimulatory interactions with hAPE1. Potential mechanisms for hAPE1 disruption of the of hTDG product complex are discussed.


Assuntos
Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/fisiologia , Timina DNA Glicosilase/metabolismo , Pareamento Incorreto de Bases , Sequência de Bases , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/química , Humanos , Cinética , Modelos Biológicos , Modelos Químicos , Conformação Molecular , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteína SUMO-1/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA