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.
Cell Mol Life Sci ; 81(1): 287, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38970705

RESUMO

Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRß, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRß as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.


Assuntos
Homeostase , Receptores X do Fígado , Macrófagos Alveolares , Pneumonia , Surfactantes Pulmonares , Transdução de Sinais , Animais , Receptores X do Fígado/metabolismo , Receptores X do Fígado/genética , Surfactantes Pulmonares/metabolismo , Camundongos , Pneumonia/metabolismo , Pneumonia/patologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pulmão/metabolismo , Pulmão/patologia , Células Epiteliais Alveolares/metabolismo , Asma/metabolismo , Asma/patologia , Asma/genética , Colesterol/metabolismo , Metabolismo dos Lipídeos , Fagocitose
2.
Adv Sci (Weinh) ; 11(20): e2307201, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38549193

RESUMO

Macrophages regulate essential aspects of innate immunity against pathogens. In response to microbial components, macrophages activate primary and secondary inflammatory gene programs crucial for host defense. The liver X receptors (LXRα, LXRß) are ligand-dependent nuclear receptors that direct gene expression important for cholesterol metabolism and inflammation, but little is known about the individual roles of LXRα and LXRß in antimicrobial responses. Here, the results demonstrate that induction of LXRα transcription by prolonged exposure to lipopolysaccharide (LPS) supports inflammatory gene expression in macrophages. LXRα transcription is induced by NF-κB and type-I interferon downstream of TLR4 activation. Moreover, LPS triggers a reprogramming of the LXRα cistrome that promotes cytokine and chemokine gene expression through direct LXRα binding to DNA consensus sequences within cis-regulatory regions including enhancers. LXRα-deficient macrophages present fewer binding of p65 NF-κB and reduced histone H3K27 acetylation at enhancers of secondary inflammatory response genes. Mice lacking LXRα in the hematopoietic compartment show impaired responses to bacterial endotoxin in peritonitis models, exhibiting reduced neutrophil infiltration and decreased expansion and inflammatory activation of recruited F4/80lo-MHC-IIhi peritoneal macrophages. Together, these results uncover a previously unrecognized function for LXRα-dependent transcriptional cis-activation of secondary inflammatory gene expression in macrophages and the host response to microbial ligands.


Assuntos
Inflamação , Receptores X do Fígado , Macrófagos , Transcriptoma , Animais , Camundongos , Modelos Animais de Doenças , Inflamação/genética , Inflamação/metabolismo , Lipopolissacarídeos , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia , Camundongos Endogâmicos C57BL
3.
Bioorg Chem ; 127: 105926, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35717804

RESUMO

Synthetic flavonoids with new substitution patterns have attracted attention as potential anticancer drugs. Here, twelve chalcones were synthesized and their antiproliferative activities against five human tumour cells were evaluated. This series of chalcone derivatives was characterized by the presence of an additional aromatic or heterocyclic ring linked by an ether, in the case of a benzyl radical, or an ester or amide functional group in the case of a furoyl radical. In addition, the influence on cytotoxicity by the presence of one or three methoxy groups or a 2,4-dimethoxy-3-methyl system on the B ring of the chalcone scaffold was also explored. The results revealed that the most cytotoxic chalcones contain a furoyl substituent linked by an ester or an amide through the 2'-hydroxy or the 2'-amino group of the A ring of the chalcone skeleton, with IC50 values between 0.2 ±â€¯0.1 µM and 1.3 ±â€¯0.1 µM against human leukaemia cells. The synthetic chalcone 2'-furoyloxy-4-methoxychalcone (FMC) was, at least, ten-fold more potent than the antineoplastic agent etoposide against U-937 cells and displayed less cytotoxicity against human peripheral blood mononuclear cells. Treatment of U-937 and HL-60 cells with FMC induced cell cycle arrest at the G2-M phase, an increase in the percentage of sub-G1 and annexin-V positive cells, the release of mitochondrial cytochrome c, activation of caspase and poly(ADP-ribose) polymerase cleavage. In addition, it inhibited tubulin polymerization in vitro in a concentration dependent manner. Cell death triggered by this chalcone was decreased by the pan-caspase inhibitor z-VAD-fmk and was dependent of the generation of reactive oxygen species. We conclude that this furoyloxychalcone may be useful in the development of a potential anti-leukaemia strategy.


Assuntos
Antineoplásicos , Chalcona , Chalconas , Leucemia , Amidas/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Chalcona/farmacologia , Chalconas/farmacologia , Chalconas/uso terapêutico , Ésteres/farmacologia , Células HL-60 , Humanos , Leucemia/metabolismo , Leucócitos Mononucleares/metabolismo , Relação Estrutura-Atividade
4.
Front Endocrinol (Lausanne) ; 12: 635923, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122329

RESUMO

Macrophages are immune cells that play crucial roles in host defense against pathogens by triggering their exceptional phagocytic and inflammatory functions. Macrophages that reside in healthy tissues also accomplish important tasks to preserve organ homeostasis, including lipid uptake/efflux or apoptotic-cell clearance. Both homeostatic and inflammatory functions of macrophages require the precise stability of lipid-rich microdomains located at the cell membrane for the initiation of downstream signaling cascades. Caveolin-1 (Cav-1) is the main protein responsible for the biogenesis of caveolae and plays an important role in vascular inflammation and atherosclerosis. The Liver X receptors (LXRs) are key transcription factors for cholesterol efflux and inflammatory gene responses in macrophages. Although the role of Cav-1 in cellular cholesterol homeostasis and vascular inflammation has been reported, the connection between LXR transcriptional activity and Cav-1 expression and function in macrophages has not been investigated. Here, using gain and loss of function approaches, we demonstrate that LXR-dependent transcriptional pathways modulate Cav-1 expression and compartmentation within the membrane during macrophage activation. As a result, Cav-1 participates in LXR-dependent cholesterol efflux and the control of inflammatory responses. Together, our data show modulation of the LXR-Cav-1 axis could be exploited to control exacerbated inflammation and cholesterol overload in the macrophage during the pathogenesis of lipid and immune disorders, such as atherosclerosis.


Assuntos
Caveolina 1/biossíntese , Colesterol/metabolismo , Receptores X do Fígado/biossíntese , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Anti-Inflamatórios , Apolipoproteína A-I/metabolismo , Aterosclerose/metabolismo , Caveolina 1/genética , Membrana Celular/metabolismo , Detergentes , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Transdução de Sinais , Transcrição Gênica
5.
Int J Radiat Oncol Biol Phys ; 104(4): 913-923, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30922944

RESUMO

PURPOSE: To evaluate the role of liver X receptor (LXR) nuclear receptors on irradiation-induced cell death and polarization of macrophages and the potential implications in the context of radiation therapy treatment of cancer. METHODS AND MATERIALS: Primary and immortalized murine bone marrow-derived macrophages (BMDMs) from wild type or LXR double knock-out mice were exposed to gamma irradiation. Subsequently, analysis of LXR signaling on cell proliferation and cytotoxicity induced by ionizing radiation was determined by time-lapse photomicroscopy. Genotoxic cell damage was evaluated by Western blot of γ-H2AX and p53. Pyroptosis was analyzed through cell viability assay, lactate dehydrogenase release assay, and Western blot of caspase-1 active protein. Expression of inflammatory markers was measured by real-time quantitative polymerase chain reaction. RESULTS: Genetic and pharmacologic inactivation of LXR induced radiosensitivity of macrophages. LXR deficiency decreased cell proliferation and enhanced cytotoxicity induced by ionizing radiation in both immortalized and primary BMDMs. Protein levels of γ-H2AX and p53, both involved in response to cell damage, were exacerbated in LXR-deficient macrophages exposed to irradiation. Cell membrane damage was augmented and cell viability was decreased in LXR-deficient macrophages compared with LXR wild type macrophages in response to irradiation. In addition, LXR deficiency enhanced both caspase-1 activation and lactate dehydrogenase release in BMDM exposed inflammasome activators. LXR inactivation or deficiency markedly increased the expression of proinflammatory markers IL-1ß, IL-6, and inducible nitric oxide synthase in irradiated macrophages. CONCLUSIONS: The present work identifies LXR transcription factors as potential therapeutic targets to enhance the suppressive effects of radiation therapy on tumor growth through induction of macrophage cell death and activation of the inflammatory cascade.


Assuntos
Sobrevivência Celular , Receptores X do Fígado/metabolismo , Macrófagos/efeitos da radiação , Tolerância a Radiação , Animais , Morte Celular , Polaridade Celular , Proliferação de Células , Quebras de DNA de Cadeia Dupla , Raios gama , Expressão Gênica , Histonas/metabolismo , Inflamação , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , L-Lactato Desidrogenase/metabolismo , Receptores X do Fígado/antagonistas & inibidores , Receptores X do Fígado/genética , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Neoplasias/radioterapia , Óxido Nítrico Sintase Tipo II/metabolismo , Piroptose , Radiação Ionizante , Reprodutibilidade dos Testes , Proteína Supressora de Tumor p53/metabolismo
6.
Methods Mol Biol ; 1951: 75-85, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30825145

RESUMO

Macrophages are professional phagocytic cells that play key roles in innate and adaptive immunity, metabolism, and tissue homeostasis. Lipid metabolism is tightly controlled at the transcriptional level, and one of the key players of this regulation in macrophages and other cell types is the LXR subfamily of nuclear receptors (LXRα and LXRß). The use of LXR double knockout (LXR-DKO) macrophages in vitro has yielded extensive benefits in metabolism research, but this technique is hindered by primary macrophage cell expansion capability, which diminishes along terminal cell differentiation process. Here we detail a method to immortalize LXR double knockout bone marrow-derived macrophage cells at an early stage of differentiation, using a retroviral delivery of a combination of murine v-myc and v-raf oncogenes. This methodology enables the generation of autonomous self-renewing macrophages bearing an LXR-DKO genetic background, as a valuable tool for research in lipid metabolism and other LXR receptor-mediated effects.


Assuntos
Receptores X do Fígado/deficiência , Macrófagos/metabolismo , Animais , Biomarcadores , Linhagem Celular Transformada , Vetores Genéticos/genética , Receptores X do Fígado/metabolismo , Macrófagos/imunologia , Camundongos , Retroviridae/genética , Transdução Genética , Transgenes
7.
Methods Mol Biol ; 1951: 99-109, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30825147

RESUMO

Liver X receptors are members of the nuclear receptor superfamily of transcription factors. The LXR genes (NR1H2 and NR1H3) encode for two different proteins referred to as LXRα and LXRß. Each LXR presents diverse tissue distribution but similar target DNA-binding elements and ligands. Both LXRs act as relevant transcriptional regulators of cholesterol metabolism in many tissues. Additionally, LXRs participate in innate immunity and inflammation. Therefore, in order to understand the molecular requirements that operate in LXR-dependent transcription, it is important to decipher LXR genomic binding properties. We have recently performed genome-wide binding analysis of LXR proteins. In this method paper, we describe a detailed computational protocol primarily based on HOMER software package for the analysis of ChIP-seq data.


Assuntos
Imunoprecipitação da Cromatina , Biologia Computacional , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Receptores X do Fígado/metabolismo , Sítios de Ligação , Imunoprecipitação da Cromatina/métodos , Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Ligação Proteica , Análise de Sequência de DNA , Software , Fatores de Transcrição/metabolismo , Navegador , Fluxo de Trabalho
8.
Mol Cell Biol ; 39(5)2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30602495

RESUMO

The liver X receptors α and ß (LXRα and LXRß) are oxysterol-activated transcription factors that coordinately regulate gene expression that is important for cholesterol and fatty acid metabolism. In addition to their roles in lipid metabolism, LXRs participate in the transcriptional regulation of macrophage activation and are considered potent regulators of inflammation. LXRs are highly similar, and despite notable exceptions, most of their reported functions are substantially overlapping. However, their individual genomic distribution and transcriptional capacities have not been characterized. Here, we report a macrophage cellular model expressing equivalent levels of tagged LXRs. Analysis of data from chromatin immunoprecipitation coupled with deep sequencing revealed that LXRα and LXRß occupy both overlapping and exclusive genomic regulatory sites of target genes and also control the transcription of a receptor-exclusive set of genes. Analysis of genomic H3K27 acetylation and mRNA transcriptional changes in response to synthetic agonist or antagonist treatments revealed a putative mode of pharmacologically independent regulation of transcription. Integration of microarray and sequencing data enabled the description of three possible mechanisms of LXR transcriptional activation. Together, these results contribute to our understanding of the common and differential genomic actions of LXRs and their impact on biological processes in macrophages.


Assuntos
Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Macrófagos/metabolismo , Animais , Proteínas de Transporte/metabolismo , Colesterol/metabolismo , Imunoprecipitação da Cromatina/métodos , Regulação da Expressão Gênica/genética , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Ativação de Macrófagos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos/genética , Regiões Promotoras Genéticas/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Ativação Transcricional
9.
Mol Carcinog ; 57(11): 1525-1539, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30035337

RESUMO

Photodynamic therapy (PDT) is a rising and hopeful treatment for solid tumors and others malignancies. PDT uses harmless visible light to activate a tumor-associated photosensitizer (PS). The excited PS generates cytotoxic reactive oxygen species (ROS) that induce damage and death of tumor cells. It is known that certain phytoalexins and phytoanticipins derived from plants often display a PS-like activity due to a phenalenone (PN) moiety-an efficient singlet oxygen photosensitizer-in its skeleton. The aim of this study is to explore the phototoxic properties of PN on the human cell line tumor-derived HL60 (acute promyelocytic leukemia) and to identify the cell-specific targets of ROS involved in the tumor cell death. Our results reveal that PN acts as an excellent PS, showing a potent antitumor cell activity in presence of light. PN-PDT generates intracellular ROS, via oxidation reaction mechanisms type I and II, resulting in an induction of apoptosis. Moreover, both extrinsic (through direct activation of caspase-3) and intrinsic (through mitochondrial depolarization) pathways of apoptosis are induced by PN-PDT. Using pharmacologic inhibitors, we also find that PN-PDT activates caspase-8/tBid and p38-MAPK, triggering the activation of the apoptotic pathways. Although, survival pathways are also promoted through PI3 K/Akt and JNK activation, the net result of PN-PDT is the tumor cell death. The present work identifies to PN, for the first time, as a potent photosensitizer in human tumor cell lines and proposes a mechanism by which ROS induces apoptosis of tumor cell.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspase 8/metabolismo , Fenalenos/farmacologia , Fotoquimioterapia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA , Relação Dose-Resposta a Droga , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estrutura Molecular , Neoplasias/genética , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
10.
Mol Cell Biol ; 38(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29507185

RESUMO

The liver X receptors (LXRs) are ligand-activated nuclear receptors with established roles in the maintenance of lipid homeostasis in multiple tissues. LXRs exert additional biological functions as negative regulators of inflammation, particularly in macrophages. However, the transcriptional responses controlled by LXRs in other myeloid cells, such as dendritic cells (DCs), are still poorly understood. Here we used gain- and loss-of-function models to characterize the impact of LXR deficiency on DC activation programs. Our results identified an LXR-dependent pathway that is important for DC chemotaxis. LXR-deficient mature DCs are defective in stimulus-induced migration in vitro and in vivo Mechanistically, we show that LXRs facilitate DC chemotactic signaling by regulating the expression of CD38, an ectoenzyme important for leukocyte trafficking. Pharmacological or genetic inactivation of CD38 activity abolished the LXR-dependent induction of DC chemotaxis. Using the low-density lipoprotein receptor-deficient (LDLR-/-) LDLR-/- mouse model of atherosclerosis, we also demonstrated that hematopoietic CD38 expression is important for the accumulation of lipid-laden myeloid cells in lesions, suggesting that CD38 is a key factor in leukocyte migration during atherogenesis. Collectively, our results demonstrate that LXRs are required for the efficient emigration of DCs in response to chemotactic signals during inflammation.


Assuntos
Quimiotaxia/fisiologia , Células Dendríticas/fisiologia , Receptores X do Fígado/fisiologia , ADP-Ribosil Ciclase 1/metabolismo , Animais , Células Cultivadas , Células Dendríticas/citologia , Inflamação , Metabolismo dos Lipídeos , Receptores X do Fígado/genética , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos , Receptores Citoplasmáticos e Nucleares , Transdução de Sinais
11.
Nat Immunol ; 14(8): 831-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770640

RESUMO

Macrophages are professional phagocytic cells that orchestrate innate immune responses and have considerable phenotypic diversity at different anatomical locations. However, the mechanisms that control the heterogeneity of tissue macrophages are not well characterized. Here we found that the nuclear receptor LXRα was essential for the differentiation of macrophages in the marginal zone (MZ) of the spleen. LXR-deficient mice were defective in the generation of MZ and metallophilic macrophages, which resulted in abnormal responses to blood-borne antigens. Myeloid-specific expression of LXRα or adoptive transfer of wild-type monocytes restored the MZ microenvironment in LXRα-deficient mice. Our results demonstrate that signaling via LXRα in myeloid cells is crucial for the generation of splenic MZ macrophages and identify an unprecedented role for a nuclear receptor in the generation of specialized macrophage subsets.


Assuntos
Hematopoese/imunologia , Macrófagos/imunologia , Receptores Nucleares Órfãos/imunologia , Baço/imunologia , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Diferenciação Celular/imunologia , Citometria de Fluxo , Imunidade Celular/imunologia , Imuno-Histoquímica , Receptores X do Fígado , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Receptores Nucleares Órfãos/agonistas , Transdução de Sinais/imunologia , Organismos Livres de Patógenos Específicos , Baço/citologia
12.
Endocrinology ; 144(11): 4763-72, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12959973

RESUMO

Systemic or intratesticular release of TNF alpha and IL1 beta have been implicated in the reduced testosterone biosynthesis and impaired production of competent spermatozoa found in human patients suffering from sepsis or chronic inflammation. Although in vitro and in vivo studies have demonstrated that TNF alpha and IL1 beta intercept the hypothalamic-pituitary testis axis at different levels, the site(s) of action and relative contribution of each cytokine to the overall testicular failure associated to systemic inflammatory processes remains poorly defined. In this study we show that intratesticular delivery of TNF alpha induced a rapid (4 h) and sustained (up to 24 h) reduction in steroidogenic acute regulatory (StAR) protein expression and testosterone biosynthesis in nonstimulated or human chorionic gonadotropin-treated intact or hypophysectomized rats. Bilateral treatment with cell-permeant short-chain ceramides (C2-cer or C6-cer) reproduced the early (4 h) inhibitory action of TNFalpha on testosterone biosynthesis and testicular StAR expression. The inhibitory action of C2-cer or C6-cer was not observed in animals treated with inactive analogs (dihydroceramide), phosphorylcholine, sphingosine, or sphingosine-1P. In sharp contrast to the previously described ability of IL1 beta to prevent human chorionic gonadotropin-stimulated Leydig cell steroidogenesis in vitro, serum testosterone and testicular StAR protein expression remained unchanged in animals bilaterally injected with this cytokine. These data support the concept that TNF alpha triggers different effector mechanisms to directly inhibit Leydig cell StAR expression and steroidogenesis, which ultimately contribute to the global reproductive failure associated with chronic inflammation and sepsis.


Assuntos
Ceramidas/administração & dosagem , Células Intersticiais do Testículo/metabolismo , Fosfoproteínas/metabolismo , Testículo/fisiologia , Testosterona/antagonistas & inibidores , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Gonadotropina Coriônica/farmacologia , Humanos , Hipofisectomia , Injeções , Interleucina-1/administração & dosagem , Isomerismo , Masculino , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/genética , RNA Mensageiro/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Testosterona/biossíntese , Testosterona/sangue , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...