Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Transplant ; 29: 963689720946277, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32841051

RESUMO

Diabetes reduces the number and induces dysfunction in circulating endothelial progenitor cells (EPCs) by mechanisms that are still uncovered. This study aims to evaluate the number, viability, phenotype, and function of EPCs in dyslipidemic mice with early diabetes mellitus and EPC infiltration in the aortic valve in order to identify possible therapeutic targets in diabetes-associated cardiovascular disease. A streptozotocin-induced diabetic apolipoprotein E knock-out (ApoE-/-) mouse model was used to identify the early and progressive changes, at 4 or 7 days on atherogenic diet after the last streptozotocin or citrate buffer injection. Blood and aortic valves from diabetic or nondiabetic ApoE-/- animals were collected.EPCs were identified as CD34 and vascular endothelial growth factor receptor 2 positive monocytes, and the expression levels of α4ß1, αVß3, αVß5, ß1, αLß2, α5 integrins, and C-X-C chemokine receptor type 4 chemokine receptor on EPC surface were assessed by flow cytometry. The number of CD34 positive cells in the aortic valve, previously found to be recruited progenitor cells, was measured by fluorescence microscopy. Our results show that aortic valves from mice fed 7 days with atherogenic diet presented a significantly higher number of CD34 positive cells compared with mice fed only 4 days with the same diet, and diabetes reversed this finding. We also show a reduction of circulatory EPC numbers in diabetic mice caused by cell senescence and lower mobilization. Dyslipidemia induced EPC death through apoptosis regardless of the presence of diabetes, as shown by the higher percent of propidium iodide positive cells and higher cleaved caspase-3 levels. EPCs from diabetic mice expressed α4ß1 and αVß3 integrins at a lower level, while the rest of the integrins tested were unaffected by diabetes or diet. In conclusion, reduced EPC number and expression of α4ß1 and αVß3 integrins on EPCs at 4 and 7 days after diabetes induction in atherosclerosis-prone mice have resulted in lower recruitment of EPCs in the aortic valve.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Dislipidemias/fisiopatologia , Células Progenitoras Endoteliais/metabolismo , Integrina alfa4beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Células-Tronco/metabolismo , Estreptozocina/uso terapêutico , Animais , Valvopatia Aórtica , Células Cultivadas , Masculino , Camundongos , Camundongos Knockout
2.
Pharmaceutics ; 12(6)2020 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-32498305

RESUMO

Calcific aortic valve disease (CAVD) is a progressive disorder that increases in prevalence with age. An important role in aortic valve calcification is played by valvular interstitial cells (VIC), that with age or in pathological conditions acquire an osteoblast-like phenotype that advances the disease. Therefore, pharmacological interventions aiming to stop or reverse the osteoblastic transition of VIC may represent a therapeutic option for CAVD. In this study, we aimed at developing a nanotherapeutic strategy able to prevent the phenotypic switch of human aortic VIC into osteoblast-like cells. We hypothesize that nanocarriers designed for silencing the Runt-related transcription factor 2 (Runx2) will stop the progress or reverse the osteodifferentiation of human VIC, induced by high glucose concentrations and pro-osteogenic factors. We report here the potential of fullerene (C60)-polyethyleneimine (PEI)/short hairpin (sh)RNA-Runx2 nano-polyplexes to efficiently down-regulate Runx2 mRNA and protein expression leading subsequently to a significant reduction in the expression of osteogenic proteins (i.e. ALP, BSP, OSP and BMP4) in osteoblast-committed VIC. The data suggest that the silencing of Runx2 could represent a novel strategy to impede the osteoblastic phenotypic shift of VIC and the ensuing progress of CAVD.

3.
Pharmaceutics ; 11(8)2019 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-31382634

RESUMO

Citrus flavonoids have well-documented protective effects on cardiovascular system, but the poor water solubility and reduced bioavailability restrict their therapeutic use. We aimed to overcome these limitations and encapsulated naringenin and hesperetin into lipid nanoemulsions (LNs), targeted to vascular cell adhesion molecule-1 (VCAM-1), which is expressed on activated endothelial cells (ECs). LNs were characterized by a hydrodynamic size of ~200 nm, negative zeta potential, an encapsulation efficiency of flavonoids higher than 80%, good in vitro stability and steady release of the cargo. The LNs were neither cytotoxic to human ECs line EA.hy926, nor provoked in vitro lysis of murine erithrocytes. Then, we tested whether these nanoformulations reduce tumor necrosis factor-alpha (TNF-α) induced EC-activation. We found that flavonoid-loaded LNs, either non-targeted or targeted to the endothelium, were taken up by the EA.hy926 cells in a dose-dependent manner, but dependent on TNF-α only in the case of endothelium-targeted LNs. Moreover, these nanoparticles inhibited both the adhesion and transmigration of THP-1 monocytes on/through activated ECs, by mechanisms involving a reduced expression of the pro-inflammatory chemokine monocyte chemotactic protein 1 (MCP-1) and diminished nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB).

4.
Toxicology ; 416: 30-43, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30711708

RESUMO

The potential human health risks following the exposure to inorganic nanoparticles (NPs) is a very important issue for their application in leather finishing industry. The aim of our study was to investigate the cytotoxic effect of silver (Ag)/titanium dioxide (TiO2) NPs on human cells. Photocatalytic NPs were prepared by electrochemical deposition of Ag on the surface of TiO2 and nitrogen (N)-TiO2 NPs and, subsequently, physico-chemical characterized. Then, a set of experiments have been performed to study the cytotoxicity and cell death mechanisms involved, the changes in cell morphology and the production of ROS induced in human keratinocytes (HaCaT) and human lung epithelial cells (A549) by exposure to NPs. Moreover, the changes in major signaling pathways and the inflammatory response induced by Ag/N-TiO2 NPs in A549 cells were investigated. The data showed that cell death by late apoptosis/necrosis is induced in cells as function of the dose and the type of NPs and is characterized by morphological changes and cytoskeletal disorganization and an increase in reactive oxygen species (ROS) production. The exposure of A549 cells to Ag/N-TiO2 NPs determine the activation of ERK1/2 MAP-kinase pathway and the release of pro-inflammatory mediators CXCL1, GM-CSF and MIF, known to be involved in the recruitment of circulating neutrophils and monocytes.


Assuntos
Queratinócitos/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Mucosa Respiratória/efeitos dos fármacos , Compostos de Prata/toxicidade , Titânio/toxicidade , Células A549 , Apoptose/efeitos dos fármacos , Catálise , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Queratinócitos/metabolismo , Queratinócitos/patologia , Necrose , Estresse Oxidativo/efeitos dos fármacos , Processos Fotoquímicos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Medição de Risco , Transdução de Sinais
5.
Pharmaceutics ; 11(1)2019 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-30669699

RESUMO

: The progress in small-interfering RNA (siRNA) therapeutics depends on the development of suitable nanocarriers to perform specific and effective delivery to dysfunctional cells. In this paper, we questioned whether P-selectin, a cell adhesion molecule specifically expressed on the surface of activated endothelial cells (EC) could be employed as a target for nanotherapeutic intervention. To this purpose, we developed and characterized P-selectin targeted PEGylated cationic liposomes able to efficiently pack siRNA and to function as efficient vectors for siRNA delivery to tumour necrosis factor-α (TNF-α) activated EC. Targeted cationic liposomes were obtained by coupling a peptide with high affinity for P-selectin to a functionalized PEGylated phospholipid inserted in the liposomes' bilayer (Psel-lipo). As control, scrambled peptide coupled cationic liposomes (Scr-lipo) were used. The lipoplexes obtained by complexation of Psel-lipo with siRNA (Psel-lipo/siRNA) were taken up specifically and at a higher extent by TNF-α activated b.End3 endothelial cells as compared to non-targeted Scr-lipo/siRNA. The Psel-lipo/siRNA delivered with high efficiency siRNA into the cells. The lipoplexes were functional as demonstrated by the down-regulation of the selected gene (GAPDH). The results demonstrate an effective targeted delivery of siRNA into cultured activated endothelial cells using P-selectin directed PEGylated cationic liposomes, which subsequently knock-down the desired gene.

6.
Int J Nanomedicine ; 13: 63-76, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29317816

RESUMO

BACKGROUND: Lipopolysaccharide (LPS) is widely recognized as a potent activator of monocytes/macrophages, and its effects include an altered production of key mediators, such as inflammatory cytokines and chemokines. The involvement of Gi protein in mediating LPS effects has been demonstrated in murine macrophages and various cell types of human origin. PURPOSE: The aim of the present work was to evaluate the potential of a Gi-protein inhibitor encapsulated in liposomes in reducing the inflammatory effects induced by LPS in monocytes/macrophages. MATERIALS AND METHODS: Guanosine 5'-O-(2-thiodiphosphate) (GOT), a guanosine diphosphate analog that completely inhibits G-protein activation by guanosine triphosphate and its analogs, was encapsulated into liposomes and tested for anti-inflammatory effects in LPS-activated THP1 monocytes or THP1-derived macrophages. The viability of monocytes/macrophages after incubation with different concentrations of free GOT or liposome-encapsulated GOT was assessed by MTT assay. MAPK activation and production of IL1ß, TNFα, IL6, and MCP1 were assessed in LPS-activated monocytes/macrophages in the presence or absence of free or encapsulated GOT. In addition, the effect of free or liposome-encapsulated GOT on LPS-stimulated monocyte adhesion to activated endothelium and on monocyte chemotaxis was evaluated. RESULTS: We report here that GOT-loaded liposomes inhibited activation of MAPK and blocked the production of the cytokines IL1ß, TNFα, IL6, and MCP1 induced by LPS in monocytes and macrophages. Moreover, GOT encapsulated in liposomes reduced monocyte adhesion and chemotaxis. All demonstrated events were in contrast with free GOT, which showed reduced or no effect on monocyte/macrophage activation with LPS. CONCLUSION: This study demonstrates the potential of liposomal GOT in blocking LPS proinflammatory effects in monocytes/macrophages.


Assuntos
Guanosina Difosfato/análogos & derivados , Inflamação/prevenção & controle , Lipossomos/administração & dosagem , Monócitos/efeitos dos fármacos , Tionucleotídeos/farmacologia , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Guanosina Difosfato/administração & dosagem , Guanosina Difosfato/farmacologia , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/toxicidade , Lipossomos/química , Lipossomos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Tionucleotídeos/administração & dosagem , Fator de Necrose Tumoral alfa
7.
Mater Sci Eng C Mater Biol Appl ; 81: 167-176, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28887961

RESUMO

Nano-hydroxyapatite (nHAp), surface functionalized with linear polyethylenimine (LPEI), was used for the preparation of biocomposites in combination with biopolymers and poly(ε-caprolactone) (PCL), by cryogelation technique, to yield biomimetic scaffolds with controlled interconnected macroporosity, mechanical stability, and predictable degradation behavior. The structural characteristics, swelling and degradation behavior of hydroxyapatite and hydroxyapatite/ß-tricalcium phosphate (ß-TCP) filled matrices were investigated as compared to the corresponding naked polymer 3D system. It was found that the homogeneity and cohesivity of the composite are significantly dependent on the size and amount of the included inorganic particles, which are thus determining the structural parameters. Surface modification with LPEI and nanodimensions favored the nHAp integration in the organic matrix, with preferential location along protein fibers, while ß-TCP microparticles induced an increased disorder in the hybrid system. The biocomposite including nHAp only was further investigated targeting biomedical uses, and proved to be non-cytotoxic and capable of acting as gene-activated matrix (GAM). It allowed sustained delivery over time (until 22days) of embedded PEI25-pDNA polyplexes at high levels of transgene expression, while insuring a decrease in cytotoxicity as compared to polyplexes alone. Experimental data recommend such biocomposite as an attractive material for regenerative medicine.


Assuntos
Nanoestruturas , Biopolímeros , Criogéis , Durapatita , Poliésteres , Polietilenoimina
8.
Mediators Inflamm ; 2016: 1625149, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27703301

RESUMO

Inflammation is a common process associated with numerous vascular pathologies. We hypothesized that targeting the inflamed endothelium by coupling a peptide with high affinity for P-selectin to the surface of dexamethasone-loaded lipid nanoemulsions will highly increase their specific binding to activated endothelial cells (EC) and reduce the cell activation. We developed and characterized dexamethasone-loaded lipid nanoemulsions directed towards P-selectin (PLN-Dex) and monitored their anti-inflammatory effects in vitro using cultured EC (EA.hy926 cells) and in vivo using a mouse model of acute inflammation [lipopolysaccharides (LPS) intravenously administered in C57BL/6 mice]. We found that PLN-Dex bound specifically to the surface of activated EC are efficiently internalized by EC and reduced the expression of proinflammatory genes, thus preventing the monocyte adhesion and transmigration to/through activated EC. Given intravenously in mice with acute inflammation, PLN-Dex accumulated at a significant high level in the lungs (compared to nontargeted nanoemulsions) and significantly reduced mRNA expression level of key proinflammatory cytokines such as IL-1ß, IL-6, and MCP-1. In conclusion, the newly developed nanoformulation, PLN-Dex, is functional in vitro and in vivo, reducing selectively the endothelium activation and the consequent monocyte infiltration and diminishing significantly the lungs' inflammation, in a mouse model of acute inflammation.


Assuntos
Dexametasona/química , Emulsões/química , Inflamação/tratamento farmacológico , Nanoestruturas/química , Selectina-P/uso terapêutico , Animais , Quimiocina CCL2/metabolismo , Emulsões/administração & dosagem , Citometria de Fluxo , Inflamação/induzido quimicamente , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanoestruturas/administração & dosagem , Selectina-P/química
9.
Cytokine ; 83: 250-261, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27180200

RESUMO

In atherosclerotic plaques, macrophages (MAC) and smooth muscle cells (SMC) frequently reside in close proximity and resistin (Rs) and fractalkine (Fk) are present at increased levels, resistin being associated with CD68 macrophages and fractalkine predominantly associated with intimal SMC; however, their role in this location is not clear, yet. The objective of this study was to determine whether the cross-talk between MAC-SMC induces changes in MAC cytokine phenotype and if Fk and Rs have a role in the process. To this purpose, macrophages (THP-1 monocytes differentiated with phorbol myristate acetate) were interacted with SMC cultured on the membrane inserts in the presence or absence of Rs or Fk. After 24h, MAC were removed from the co-culture and the gene and protein expression of 57 cytokines was assessed by QPCR and Proteome Profiler™ Array. Fk secreted in the culture medium following MAC-SMC interaction was determined (ELISA assay) and the role of Fk in MAC cytokine gene expression was assessed by silencing the Fk receptor in both cell types. The results showed that subsequent to the interaction with SMC, MAC exhibit: (1) a general increased expression of chemokines (the highest fold increase: VCC-1 and GRO-α) and of some interleukins, such as interleukins IL-5 (∼8-fold) and IL-6; (2) an increased Fk expression that in turn induces expression of: CXCL17, CCL19, CCL2, CXCL10, CXCL12, CXCL4, CXCL7, CCL4, CCL18, CXCL16, CXCL1 and IL-27; (3) in the presence of Rs, a predominant increased expression of interleukins (the highest fold increase: IL-6, IL-27, IL-23 and IL-5) and an augmented expression of some chemokines such as MIP-1ß, GRO-α and CCL1. In addition, the secretome collected from the SMC-MAC co-culture increased human monocytes chemotaxis. DAVID analysis of the data revealed that the switch of MAC to a pro-inflammatory phenotype, prime the cells to intervene in the immune response, chemotaxis and inflammatory response. In conclusion, MAC cytokines expression is considerable augmented upon their interaction with SMC and Fk and Rs have distinct immunomodulatory roles: Fk predominantly increases the pro-angiogenic and inflammatory chemokines expression and Rs mostly the pro-inflammatory interleukins with consequences on monocyte chemotaxis. The novel data could help to develop targeted nanotherapies to reduce leukocyte chemotaxis and the ensuing inflammatory process associated with atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Quimiocina CX3CL1/metabolismo , Quimiotaxia , Regulação da Expressão Gênica , Macrófagos/metabolismo , Miócitos de Músculo Liso/metabolismo , Resistina/metabolismo , Aterosclerose/patologia , Linhagem Celular Tumoral , Humanos , Macrófagos/patologia , Miócitos de Músculo Liso/patologia
10.
J Pharm Pharmacol ; 68(2): 195-207, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26748549

RESUMO

OBJECTIVES: To prepare and characterize in vitro and in vivo lipid nanoemulsions (LN) loaded with curcumin (Cm) and functionalized with a cell-penetrating peptide. METHODS: Curcumin-loaded lipid nanoemulsions (CmLN) functionalized with a nona-arginine peptide (R9-CmLN) have been obtained, characterized and optimized for size, entrapment efficiency and in vitro Cm release. The interaction of R9-CmLN with human endothelial cells (HEC) was investigated using cultured EA.hy926 cells, and in vivo biodistribution studies were performed using C57BL6 mice. KEY FINDINGS: When used in therapeutically relevant concentration, R9-CmLN have low haemolytic activity, low cytotoxicity on HEC, and show anti-inflammatory effects by reducing the monocytes adhesion to TNF-α activated HEC. Moreover, HEC uptake and internalization of R9-CmLN was significantly higher compared to the non-functionalized CmLN. In vivo biodistribution studies in mice revealed a higher accumulation of R9-CmLN in the liver and the lungs compared to CmLN and the body clearance of the both nanoformulations after 72 h. CONCLUSIONS: Cell-penetrating peptides-functionalized CmLN have superior characteristics compared to their non-functionalized counterparts: are more efficiently internalized by the cells, produces anti-inflammatory effects in HEC and when administrated intravenously in mice exhibit increased accumulation in the liver and the lungs, suggesting their potential therapeutic applications in different inflammatory pathologies localized in the liver or the lungs.


Assuntos
Anti-Inflamatórios não Esteroides/química , Peptídeos Penetradores de Células/química , Curcumina/química , Portadores de Fármacos/química , Células Endoteliais/efeitos dos fármacos , Nanoestruturas/química , Fosfatidilcolinas/química , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Curcumina/administração & dosagem , Curcumina/farmacocinética , Curcumina/farmacologia , Composição de Medicamentos , Emulsões , Células Endoteliais/imunologia , Humanos , Camundongos Endogâmicos C57BL
11.
ACS Biomater Sci Eng ; 2(1): 104-111, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33418648

RESUMO

The design of performant nonviral vectors for efficient cellular DNA uptake represents a grand challenge. The variability of DNA targets and of the transfected cells limits the discovery of highly active nonviral vectors. Dynamic constitutional strategy presented here, combining easy synthesis and rapid screening, enables the selection of dynameric frameworks, DFs, for DNA transfection. On the basis of reversible recombination of cationic heads and of hydrophilic/hydrophobic network-constitutive building blocks, the multivalent core-shell dendritic architectures with an optimal diameter of 100 nm may be adaptively generated in the presence of DNA targets. The fittest DFs simultaneously exhibit optimal DNA binding, superior transfection yield to standard transfection SuperFect agent and preserve high HEK 293T cell viability. The present results constitutes an important advancement toward novel biologically friendly, low-cost, and efficient nonviral vectors.

12.
J Mater Chem B ; 3(42): 8250-8267, 2015 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-32262881

RESUMO

Transfection of nucleic acid molecules, large enough to interfere with the genetic mechanisms of active cells, can be performed by means of small carriers, able to collectively collaborate in generating cargocomplexes that could be involved in passive mechanisms of cellular uptake. The present work describes the synthesis, characterization, and evaluation of transfection efficacy of a conjugate molecule, which comprises a cyclic siloxane ring (2,4,6,8-tetramethylcyclotetrasiloxane, cD4 H) as the core, and, on average, 3.76 molecules of 2 kDa polyethyleneimine (PEI) as cationic branches, with an average molecular mass of 7.3 kDa. As demonstrated by in silico molecular modeling and dynamic simulation, the conjugate molecule (cD4 H-AGE-PEI) tends to adopt an asymmetric structure, specific for amphipathic molecules (confirmed by a log P value of -1.902 ± 0.06), that favors a rapid interaction with nucleic acids. The conjugate and the polyplexes with the pEYFP plasmid were proved to be non-cytotoxic, and capable of ensuring transfection yields better than 30%, on HEK 293T cell culture, superior to the value obtained using the SuperFect® reagent. We presume that the increased transfection efficacy originates in the ability of the conjugate to locally tightly encompass pDNA molecules by electrostatic interaction mediated by the short PEI branches, and consequently to expose the siloxane hydrophobic moiety, which decreases the interaction energy with the lipid layers.

13.
Eur J Pharm Biopharm ; 89: 18-29, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25438248

RESUMO

Chemokines are critically involved in the development of chronic inflammatory-associated diseases such as atherosclerosis. We hypothesized that targeted delivery of compounds to the surface of activated endothelial cells (EC) interferes with chemokine/receptor interaction and thereby efficiently blocks inflammation. We developed PEGylated target-sensitive liposomes (TSL) encapsulating a CCR2 antagonist (Teijin compound 1) coupled with a specific peptide recognized by endothelial VCAM-1 (Vp-TSL-Tj). TSL were characterized for size (by dynamic light scattering), the amount of peptide coupled at the liposomal surface and Teijin release (by HPLC). We report that Vp-TSL-Tj binds specifically to activated EC in vitro and in situ, release the entrapped Teijin and prevent the transmigration of monocytes through activated EC. This is the first evidence that nanocarriers which transport and release chemokine inhibitors at specific pathological sites can reduce chemokine-dependent inflammatory processes.


Assuntos
Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Lipossomos/farmacologia , Monócitos/efeitos dos fármacos , Receptores CCR2/antagonistas & inibidores , Molécula 1 de Adesão de Célula Vascular/metabolismo , Benzamidas/farmacologia , Células Cultivadas , Quimiocinas/antagonistas & inibidores , Portadores de Fármacos/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Monócitos/metabolismo , Nanopartículas/administração & dosagem , Pirrolidinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...