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










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 9(11): e112010, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25369374

RESUMEN

Toll-like receptor signaling, mediated by functional Toll/interleukin-1 receptor (TIR) domains, plays a critical role in activating the innate immune response responsible for controlling and clearing infection. Bacterial protein mimics of components of this signaling pathway have been identified and function through inhibition of interactions between Toll-like receptors (TLRs) and their adaptor proteins, mediated by TIR domains. A previously uncharacterized gene, which we have named tcpF (for TIR domain-containing protein in E. faecalis) was identified in the genome of Enterococcus faecalis V583, and predicted to encode a protein resembling mammalian and bacterial TIR proteins. We overexpressed and purified TcpF from E. coli and found that the recombinant protein could bind to phosphatidylinositol phosphates in vitro, suggesting a mechanism by which TcpF may be anchored to the plasma membrane in close proximity to TIR domains of TLRs and adaptor proteins. Purified TcpF was also found to interact specifically with the TIR adaptor protein MyD88, and this interaction was dependent on the BB loop domain in the Box 2 region of TcpF. Despite no evidence of TcpF being a secreted protein, recombinant TcpF was effectively able to enter RAW264.7 cells in vitro although the mechanism by which this occurs remains to be determined. Overexpression of TcpF in mammalian cells suppressed the NF-κB activation induced by bacterial lipoteichoic acid. A mutant lacking the tcpF gene was attenuated for survival in macrophages, with increased ability to activate NF-κB compared to the wild type strain. Complementation in trans restored growth, and inhibition of NF-κB, to that of wild type levels. No appreciable difference in bacterial persistence, dissemination or pathogenesis was observed between the wild type and mutant in a mouse peritonitis model however, which suggested either a subtle role for TcpF or functional overlap with other redundant factor(s) in this virulence model.


Asunto(s)
Proteínas Bacterianas/fisiología , Enterococcus faecalis/fisiología , Factor 88 de Diferenciación Mieloide/fisiología , FN-kappa B/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Línea Celular , Femenino , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Interacciones Huésped-Patógeno , Lipopolisacáridos/fisiología , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Datos de Secuencia Molecular , Peritonitis/metabolismo , Peritonitis/microbiología , Fagocitosis , Estructura Terciaria de Proteína , Transducción de Señal , Ácidos Teicoicos
2.
FEMS Immunol Med Microbiol ; 65(2): 270-82, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22333034

RESUMEN

Enterococcus faecalis is a commensal organism of the gastrointestinal tract but can also cause serious opportunistic infections. In addition to high levels of antibiotic resistance, the ability to form biofilms on abiotic surfaces and on in-dwelling devices within the host complicates treatment strategies and successful outcomes of antibiotic therapy. Despite rapid advances made in recent years in understanding the genomics and virulence of this organism, much remains to be learned regarding the host response to enterococcal infections. In this study, we investigated the interaction of RAW264.7 macrophages and JAWS II dendritic cells with biofilm and planktonic E. faecalis, in vitro. Specifically, we compared phagocytosis, intracellular survival, secretion of proinflammatory cytokines, and the activation and maturation of phagocytes. Our results revealed that both macrophages and dendritic cells phagocytize biofilm mode cells at levels equal to or better than their planktonic counterparts. Internalized biofilm bacteria showed relatively greater survival at 24 h in macrophages than in dendritic cells and led to slightly higher expression of phagocyte activation markers. Macrophages infected with biofilm cells also secreted lower levels of proinflammatory cytokines studied. Overall, these results suggest that biofilm E. faecalis may be better adapted to overcome host defenses in vivo.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Enterococcus faecalis/inmunología , Enterococcus faecalis/fisiología , Macrófagos/inmunología , Macrófagos/microbiología , Animales , Línea Celular , Citocinas/metabolismo , Ratones , Viabilidad Microbiana
3.
Plasmid ; 64(1): 18-25, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20307569

RESUMEN

Enterococcus faecalis has emerged as a prominent healthcare-associated pathogen frequently encountered in bacteremia, endocarditis, urinary tract infection, and as a leading cause of antibiotic-resistant infections. We recently demonstrated a capacity for high-level biofilm formation by a clinical E. faecalis isolate, E99. This high biofilm-forming phenotype was attributable to a novel locus, designated bee, specifying a pilus at the bacterial cell surface and localized to a large approximately 80 kb conjugative plasmid. To better understand the origin of the bee locus, as well as to potentially identify additional factors important to the biology and pathogenesis of strain E99, we sequenced the entire plasmid. The nucleotide sequence of the plasmid, designated pBEE99, revealed large regions of identity to the previously characterized conjugative plasmid pCF10. In addition to the bee locus, pBEE99 possesses an open reading frame potentially encoding aggregation substance, as well as open reading frames putatively encoding polypeptides with 60% to 99% identity at the amino acid level to proteins involved in regulation of the pheromone response and conjugal transfer of pCF10. However, strain E99 did not respond to the cCF10 pheromone in clumping assays. While pBEE99 was found to be devoid of any readily recognizable antibiotic resistance determinants, it carries two non-identical impB/mucB/samB-type genes, as well as genes potentially encoding a two-component bacteriocin similar to that encoded on pYI14. Although no bacteriocin activity was detected from an OG1RF transconjugant carrying pBEE99 against strain FA2-2, it was approximately an order of magnitude more resistant to ultraviolet radiation. Moreover, curing strain E99 of this plasmid significantly reduced its ability to survive UV exposure. Therefore, pBEE99 represents a novel conjugative plasmid that confers biofilm-forming and enhanced UV resistance traits that might potentially impact the virulence and/or fitness of E. faecalis.


Asunto(s)
Conjugación Genética/efectos de la radiación , Enterococcus faecalis/genética , Enterococcus faecalis/efectos de la radiación , Plásmidos/genética , Tolerancia a Radiación/genética , Rayos Ultravioleta , Bacteriocinas/farmacología , Secuencia de Bases , Conjugación Genética/efectos de los fármacos , Enterococcus faecalis/efectos de los fármacos , Oligopéptidos/genética , Sistemas de Lectura Abierta/genética , Feromonas/genética , Mapeo Físico de Cromosoma , Tolerancia a Radiación/efectos de la radiación
4.
J Bacteriol ; 191(10): 3392-402, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19270086

RESUMEN

Enterococcus faecalis is a leading cause of nosocomial infections and is known for its ability to acquire and transfer virulence and antibiotic resistance determinants from other organisms. A 150-kb pathogenicity island (PAI) encoding several genes that contribute to pathogenesis was identified among antibiotic-resistant clinical isolates. In the current study, we examined the structure of the PAI in a collection of isolates from diverse sources in order to gain insight into its genesis and dynamics. Using multilocus sequence typing to assess relatedness at the level of strain background and microarray analysis to identify variations in the PAI, we determined the extent to which structural variations occur within the PAI and also the extent to which these variations occur independently of the chromosome. Our findings provide evidence for a modular gain of defined gene clusters by the PAI. These results support horizontal transfer as the mechanism for accretion of genes into the PAI and highlight a likely role for mobile elements in the evolution of the E. faecalis PAI.


Asunto(s)
Enterococcus faecalis/genética , Evolución Molecular , Variación Genética/genética , Islas Genómicas/genética , Enterococcus faecalis/clasificación , Transferencia de Gen Horizontal/genética , Familia de Multigenes/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia
5.
Infect Immun ; 76(12): 5668-76, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18824537

RESUMEN

A gene encoding a putative AraC-type transcriptional regulator was identified on the 153-kb pathogenicity island (PAI) found among virulent Enterococcus faecalis strains. In an effort to understand the function of this regulator, designated PerA (for pathogenicity island-encoded regulator), we first examined the expression of the perA gene in the original PAI strain MMH594 and in an unrelated clinical isolate E99 by reverse transcription-PCR. Interestingly, expression analysis revealed no detectable perA transcript in MMH594, whereas a transcript was observed in strain E99. Nucleotide sequence analysis revealed that this altered expression between the two strains was attributable to the differential location of an IS1191 element within the putative promoter region upstream of the perA gene. In order to determine the role of this putative regulator in E. faecalis pathogenesis, a perA-deficient mutant was created in strain E99, and the wild-type and mutant pair were compared for phenotypic differences. In in vitro biofilm assays, the mutant strain showed a significantly higher level of growth medium-specific biofilm formation compared to the wild type. However, in a murine intraperitoneal infection model, the mutant strain was significantly less pathogenic. The mutant was also attenuated for survival within macrophages in vitro. These findings highlight the importance of PerA as a regulator of biofilm formation and survival within macrophages and is likely a regulator controlling determinants important to pathogenesis.


Asunto(s)
Factor de Transcripción de AraC/genética , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidad , Islas Genómicas/genética , Infecciones por Bacterias Grampositivas/genética , Animales , Secuencia de Bases , Biopelículas , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Macrófagos/microbiología , Macrófagos/ultraestructura , Ratones , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
BMC Bioinformatics ; 8 Suppl 7: S2, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18047719

RESUMEN

BACKGROUND: To better understand the response of urinary epithelial (urothelial) cells to Enterococcus faecalis, a uropathogen that exhibits resistance to multiple antibiotics, a genome-wide scan of gene expression was obtained as a time series from urothelial cells growing as a layered 3-dimensional culture similar to normal urothelium. We herein describe a novel means of analysis that is based on deconvolution of gene variability into technical and biological components. RESULTS: Analysis of the expression of 21,521 genes from 30 minutes to 10 hours post infection, showed 9553 genes were expressed 3 standard deviations (SD) above the system zero-point noise in at least 1 time point. The asymmetric distribution of relative variances of the expressed genes was deconvoluted into technical variation (with a 6.5% relative SD) and biological variation components (>3 SD above the mode technical variability). These 1409 hypervariable (HV) genes encapsulated the effect of infection on gene expression. Pathway analysis of the HV genes revealed an orchestrated response to infection in which early events included initiation of immune response, cytoskeletal rearrangement and cell signaling followed at the end by apoptosis and shutting down cell metabolism. The number of poorly annotated genes in the earliest time points suggests heretofore unknown processes likely also are involved. CONCLUSION: Enterococcus infection produced an orchestrated response by the host cells involving several pathways and transcription factors that potentially drive these pathways. The early time points potentially identify novel targets for enhancing the host response. These approaches combine rigorous statistical principles with a biological context and are readily applied by biologists.


Asunto(s)
Mapeo Cromosómico/métodos , Enterococcus faecalis/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Proteoma/fisiología , Urotelio/metabolismo , Urotelio/microbiología , Células Cultivadas , Células Epiteliales/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Variación Genética/genética , Humanos , Biología de Sistemas/métodos , Urotelio/citología
7.
Mol Microbiol ; 63(2): 530-44, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17163979

RESUMEN

Enterococcus faecalis, a leading cause of nosocomial antibiotic resistant infections, frequently possesses a 150 kb pathogenicity island (PAI) that carries virulence determinants. The presence of excisionase and integrase genes, conjugative functions and multiple insertion sequence elements suggests that the PAI, or segments thereof, might be capable of horizontal transfer. In this report, the transfer of the E. faecalis PAI is demonstrated and a mechanism for transfer elucidated. In filter matings, chloramphenicol resistance was observed to transfer from strain MMH594b, a clinical isolate possessing the PAI tagged with a cat marker, to OG1RF (pCGC) with a frequency of 3.2 x 10(-10) per donor. Secondary transfer from primary transconjugant TCRFB1 to strain JH2SS in filter and broth matings occurred with a frequency of 1 and 2 x 10(-1) per donor respectively. Analysis of the transconjugants demonstrated that a 27,744 bp internal PAI segment was capable of excision and circularization in the donor, and is mobilized as a cointegrate with a pTEF1-like plasmid. High-frequency transfer also occurred from TCRFB1 to JH2SS during transient colonization of the mouse gastrointestinal tract. This is the first demonstration of the horizontal transfer of PAI-encoded virulence determinants in E. faecalis and has implications for genome evolution and diversity.


Asunto(s)
Proteínas Bacterianas/genética , Enterococcus faecalis/genética , Transferencia de Gen Horizontal , Islas Genómicas/genética , Factores de Virulencia/genética , Animales , Resistencia al Cloranfenicol/genética , Conjugación Genética , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Enterococcus faecalis/fisiología , Tracto Gastrointestinal/microbiología , Genes Reporteros , Ratones , Modelos Animales , Plásmidos/genética , Recombinación Genética , Estómago/microbiología
8.
J Clin Microbiol ; 44(11): 4200-3, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16957034

RESUMEN

Enterococcus faecalis isolates of porcine origin were screened for the presence of a previously identified pathogenicity island (PAI). By using the esp gene as a genetic marker for the presence of this PAI, 9 esp-positive and 10 esp-negative isolates of porcine origin were investigated by use of a designed oligonucleotide array. The results indicated the clustering of esp-positive strains by multilocus sequence typing (MLST), but surprisingly, all strains investigated contained parts of the PAI. None of the strains of animal origin investigated belonged to previously identified MLST complex 2, where most isolates from patients cluster. Five of the nine esp-positive E. faecalis isolates of animal origin belonged to the same PAI complex as human isolate MMH594 but differed in their sequence types, which strongly indicates the horizontal transfer of the PAI between enterococci of porcine and human origin.


Asunto(s)
Enterococcus faecalis/genética , Islas Genómicas/genética , Porcinos/microbiología , Animales , Técnicas de Tipificación Bacteriana , Enterococcus faecalis/clasificación , Enterococcus faecalis/patogenicidad , Humanos , Análisis de Secuencia de ADN
9.
J Bacteriol ; 188(6): 2063-72, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16513736

RESUMEN

Enterococci are opportunistic pathogens and among the leading causes of nosocomial infections. Enterococcus faecalis, the dominant species among infection-derived isolates, has recently been recognized as capable of forming biofilms on abiotic surfaces in vitro as well as on indwelling medical devices. A few bacterial factors known to contribute to biofilm formation in E. faecalis have been characterized. To identify additional factors which may be important to this process, we utilized a Tn917-based insertional mutagenesis strategy to generate a mutant bank in a high-biofilm-forming E. faecalis strain, E99. The resulting mutant bank was screened for mutants exhibiting a significantly reduced ability to form biofilms. One mutant, P101D12, which showed greater than 70% reduction in its ability to form biofilms compared to the wild-type parent, was further characterized. The single Tn917 insertion in P101D12 was mapped to a gene, bee-2, encoding a probable cell wall-anchored protein. Sequence information for the region flanking bee-2 revealed that this gene was a member of a locus (termed the bee locus for biofilm enhancer in enterococcus) comprised of five genes encoding three putative cell wall-anchored proteins and two probable sortases. Contour-clamped homogeneous electric field gel and Southern hybridization analyses suggested that the bee locus is likely harbored on a large conjugative plasmid. Filter mating assays using wild-type E99 or mutant P101D12 as a donor confirmed that the bee locus could transfer conjugally at high frequency to recipient E. faecalis strains. This represents the first instance of the identification of a mobile genetic element conferring biofilm-forming property in E. faecalis.


Asunto(s)
Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Enterococcus faecalis/fisiología , Proteínas de la Membrana/fisiología , Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Southern Blotting , Mapeo Cromosómico , Conjugación Genética , Cisteína Endopeptidasas , Elementos Transponibles de ADN , ADN Bacteriano/química , ADN Bacteriano/genética , Electroforesis en Gel de Agar , Enterococcus faecalis/genética , Transferencia de Gen Horizontal , Genes Bacterianos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Mutagénesis Insercional , Mutación , Fenotipo , Plásmidos/genética , Homología de Secuencia de Aminoácido
10.
J Bacteriol ; 187(17): 6213-22, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16109963

RESUMEN

Enterococci have emerged as one of the leading causes of nosocomial bloodstream, surgical site, and urinary tract infections. More recently, enterococci have been associated with biofilms, which are bacterial communities attached to a surface and encased in an extracellular polymeric matrix. The enterococcal cell surface-associated protein, Esp, enhances biofilm formation by Enterococcus faecalis in a glucose-dependent manner. Mature Esp consists of a nonrepeat N-terminal domain and a central region made up of two types of tandem repeats followed by a C-terminal membrane-spanning and anchor domain. This study was undertaken to localize the specific domain(s) of Esp that plays a role in Esp-mediated biofilm enhancement. To achieve this objective, we constructed in-frame deletion mutants expressing truncated forms of Esp in an isogenic background. By comparing strains expressing the mutant forms of Esp to those expressing wild-type Esp, we found that the strain expressing Esp lacking the N-terminal domain formed biofilms that were quantitatively less in biovolume than the strain expressing wild-type Esp. Furthermore, an E. faecalis strain expressing only the N-terminal domain of Esp fused to a heterologous protein anchor formed biofilms that were quantitatively similar to those formed by a strain expressing full-length Esp. This suggested that the minimal region contributing to Esp-mediated biofilm enhancement in E. faecalis was confined to the nonrepeat N-terminal domain. Expression of full-length E. faecalis Esp in heterologous host systems of esp-deficient Lactococcus lactis and Enterococcus faecium did not enhance biofilm formation as was observed for E. faecalis. These results suggest that Esp may require interaction with an additional E. faecalis-specific factor(s) to result in biofilm enhancement.


Asunto(s)
Proteínas Bacterianas/química , Enterococcus faecalis/metabolismo , Proteínas de la Membrana/química , Proteínas Bacterianas/genética , Biopelículas , Enterococcus faecalis/genética , Enterococcus faecalis/ultraestructura , Proteínas de la Membrana/genética , Microscopía Confocal , Mutagénesis , Plásmidos , Proteínas Recombinantes/química , Eliminación de Secuencia
11.
FEMS Microbiol Lett ; 242(2): 217-9, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15621440

RESUMEN

Enterococcal surface protein (Esp) is a cell wall-associated protein of Enterococcus faecalis that has been identified as a potential virulence factor. We used a mouse model to examine whether Esp facilitates intestinal colonization or translocation of E. faecalis to mesenteric lymph nodes. After clindamycin treatment, similar levels of high-density colonization were established after orogastric inoculation of an E. faecalis isolate containing the esp gene within a large pathogenicity island and an isogenic mutant created by allelic replacement of the esp gene with a chloramphenicol resistance cassette (P=0.7); translocation to mesenteric lymph nodes was detected in 3 of 12 (25%) mice in both groups. Isogenic mutants of FA2-2 (a plasmid-free derivative of E. faecalis strain JH2) with or without the esp gene failed to establish colonization of clindamycin-treated mice. These results suggest that Esp does not facilitate intestinal colonization or translocation of E. faecalis.


Asunto(s)
Proteínas Bacterianas/fisiología , Clindamicina/farmacología , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecalis/patogenicidad , Ganglios Linfáticos/microbiología , Proteínas de la Membrana/fisiología , Animales , Clindamicina/uso terapéutico , Modelos Animales de Enfermedad , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/genética , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Ratones
12.
Infect Immun ; 72(10): 6032-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15385507

RESUMEN

Enterococci play a dual role in human ecology. They serve as commensal organisms of the gastrointestinal tract and are also leading causes of multiple antibiotic-resistant hospital-acquired infection. Many nosocomial infections result from the ability of microorganisms to form biofilms. The molecular mechanisms involved in enterococcal biofilm formation are only now beginning to be understood. Enterococcal surface protein, Esp, has been reported to contribute to biofilm formation by Enterococcus faecalis. Recent studies have shown that enterococci form biofilms independently of Esp expression. To precisely determine what role Esp plays in E. faecalis biofilm formation, Esp was expressed on the cell surface of genetically well-defined, natively Esp-deficient strains, and isogenic Esp-positive and Esp-deficient strains were compared for their biofilm-forming ability. The results show that Esp expression leads to a significant increase in biofilm formation, irrespective of the strain tested. The contribution of Esp to biofilm formation was found to be most pronounced in the presence of 0.5% (wt/vol) or greater glucose. These results unambiguously define Esp as a key contributor to the ability of E. faecalis to form biofilms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecalis/metabolismo , Proteínas de la Membrana/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Enterococcus faecalis/química , Enterococcus faecalis/genética , Ensayo de Inmunoadsorción Enzimática , Gelatinasas/metabolismo , Glucosa/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Polipropilenos/metabolismo , Cloruro de Polivinilo/metabolismo
13.
Nature ; 417(6890): 746-50, 2002 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12066186

RESUMEN

Enterococci are members of the healthy human intestinal flora, but are also leading causes of highly antibiotic-resistant, hospital-acquired infection. We examined the genomes of a strain of Enterococcus faecalis that caused an infectious outbreak in a hospital ward in the mid-1980s (ref. 2), and a strain that was identified as the first vancomycin-resistant isolate in the United States, and found that virulence determinants were clustered on a large pathogenicity island, a genetic element previously unknown in this genus. The pathogenicity island, which varies only subtly between strains, is approximately 150 kilobases in size, has a lower G + C content than the rest of the genome, and is flanked by terminal repeats. Here we show that subtle variations within the structure of the pathogenicity island enable strains harbouring the element to modulate virulence, and that these variations occur at high frequency. Moreover, the enterococcal pathogenicity island, in addition to coding for most known auxiliary traits that enhance virulence of the organism, includes a number of additional, previously unstudied genes that are rare in non-infection-derived isolates, identifying a class of new targets associated with disease which are not essential for the commensal behaviour of the organism.


Asunto(s)
Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidad , Resistencia a la Vancomicina/genética , Composición de Base , Secuencia de Bases , Infección Hospitalaria/microbiología , Genes Bacterianos/genética , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia/genética , Estados Unidos , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...