Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Virology ; 587: 109871, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37634292

RESUMEN

Mammalian orthoreovirus (MRV) is an oncolytic virus that has been tested in over 30 clinical trials. Increased clinical success has been achieved when MRV is used in combination with other onco-immunotherapies. This has led the field to explore the creation of recombinant MRVs which incorporate immunotherapeutic sequences into the virus genome. This work focuses on creation and characterization of a recombinant MRV, S1/HER2nhd, which encodes a truncated σ1 protein fused in frame with three human epidermal growth factor receptor 2 (HER2) peptides (E75, AE36, and GP2) known to induce HER2 specific CD8+ and CD4+ T cells. We show S1/HER2nhd expresses the σ1 fusion protein containing HER2 peptides in infected cells and on the virion, and infects, replicates in, and reduces survival of HER2+ breast cancer cells. The oncolytic properties of MRV combined with HER2 peptide expression holds potential as a vaccine to prevent recurrences of HER2 expressing cancers.


Asunto(s)
Neoplasias , Orthoreovirus de los Mamíferos , Animales , Humanos , Orthoreovirus de los Mamíferos/genética , Proteínas Recombinantes de Fusión/genética , Péptidos , Mamíferos
2.
bioRxiv ; 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37214868

RESUMEN

Mammalian orthoreovirus (MRV) is a clinically benign oncolytic virus which has been investigated for use in multiple cancer types, including breast cancer (BC). In human clinical trials, MRV has been shown to be safe, and multiple BC patients have shown partial responses to intratumoral and intravenous virus delivery. Combination therapies inclusive of MRV and current FDA approved BC chemotherapies are being investigated to target metastatic, early BC, and triple negative BC. Though MRV is being tested clinically, we still do not fully understand the highly variable patient responses to MRV therapy. One of the most aggressive BC subtypes is HER2+ BC, in which human epidermal growth factor receptor 2 (HER2) is dysregulated, resulting in increased growth, survival, and metastasis of cancer cells. FDA approved therapies, trastuzumab and pertuzumab, target HER2 to prevent signaling of the phosphoinositide 3-kinase (PI3K) pathway. However, recent findings show that accumulation of hypoxia inducible factor-1 alpha (HIF-1α) in HER2+ BC cells contributes to trastuzumab resistance. In this work, we provide evidence that MRV infects, replicates in, and kills HER2 overexpressing cells. MRV infection is also found to have variable effects on signaling pathways that activate or are activated by HER2 expression. Finally, we show that MRV reduces HIF-1α accumulation in all the cell lines tested, including a HER2+ BC cell line. These studies provide further evidence that MRV holds promise for use in conjunction with trastuzumab to treat HER2+ BC patients.

3.
Virology ; 558: 38-48, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33721728

RESUMEN

Mammalian orthoreovirus (MRV) is a safe and effective cancer killing virus that has completed Phase I-III clinical trials against numerous cancer types. While many patients experience benefit from MRV therapy, pre-defined set points necessary for FDA approval have not been reached. Therefore, additional research into MRV biology and the effect of viral therapy on different tumor genetic subtypes and microenvironments is necessary to identify tumors most amenable to MRV virotherapy. In this work we analyzed the stage of viral infection necessary to inhibit HIF-1α, an aggressive cancer activator induced by hypoxia. We demonstrated that two viral capsid proteins were not necessary and that a step parallel with virus core movement across the endosomal membrane was required for this inhibition. Altogether, this work clarifies the mechanisms of MRV-induced HIF-1α inhibition and provides biological relevance for using MRV to inhibit the devastating effects of tumor hypoxia.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Viroterapia Oncolítica , Orthoreovirus de los Mamíferos/fisiología , Neoplasias de la Próstata , Microambiente Tumoral/fisiología , Línea Celular Tumoral , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Masculino , Hipoxia Tumoral
4.
Viruses ; 13(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525628

RESUMEN

Cells are continually exposed to stressful events, which are overcome by the activation of a number of genetic pathways. The integrated stress response (ISR) is a large component of the overall cellular response to stress, which ultimately functions through the phosphorylation of the alpha subunit of eukaryotic initiation factor-2 (eIF2α) to inhibit the energy-taxing process of translation. This response is instrumental in the inhibition of viral infection and contributes to evolution in viruses. Mammalian orthoreovirus (MRV), an oncolytic virus that has shown promise in over 30 phase I-III clinical trials, has been shown to induce multiple arms within the ISR pathway, but it successfully evades, modulates, or subverts each cellular attempt to inhibit viral translation. MRV has not yet received Food and Drug Administration (FDA) approval for general use in the clinic; therefore, researchers continue to study virus interactions with host cells to identify circumstances where MRV effectiveness in tumor killing can be improved. In this review, we will discuss the ISR, MRV modulation of the ISR, and discuss ways in which MRV interaction with the ISR may increase the effectiveness of cancer therapeutics whose modes of action are altered by the ISR.


Asunto(s)
Virus Oncolíticos/fisiología , Orthoreovirus de los Mamíferos/fisiología , Infecciones por Reoviridae/virología , Estrés Fisiológico , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Viroterapia Oncolítica , Fosforilación , Biosíntesis de Proteínas , Caperuzas de ARN/metabolismo , Infecciones por Reoviridae/metabolismo
5.
Virology ; 485: 153-61, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26264969

RESUMEN

As prostate tumor cell growth depends on hormones, androgen ablation is an effective therapy for prostate cancer (PCa). However, progression of PCa cells to androgen independent growth (castrate resistant prostate cancer, CRPC) results in relapse and mortality. Hypoxia, a microenvironment of low oxygen that modifies the activity of PCa regulatory proteins including the androgen receptor (AR), plays a critical role in progression to CRPC. Therapies targeting hypoxia and the AR may lengthen the time to CRPC progression thereby increasing survival time of PCa patients. Mammalian Orthoreovirus (MRV) has shown promise for the treatment of prostate tumors in vitro and in vivo. In this study, we found that MRV infection induces downregulation of proteins implicated in CRPC progression, interferes with hypoxia-induced AR activity, and induces apoptosis in androgen dependent cells. This suggests MRV possesses traits that could be exploited to create novel therapies for the inhibition of progression to CRPC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Virus Oncolíticos/genética , Orthoreovirus de los Mamíferos/genética , Próstata/virología , Receptores Androgénicos/genética , Andrógenos/metabolismo , Apoptosis/genética , Hipoxia de la Célula , Línea Celular Tumoral , Humanos , Calicreínas/genética , Calicreínas/metabolismo , Masculino , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Orthoreovirus de los Mamíferos/metabolismo , Próstata/metabolismo , Próstata/patología , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal
6.
Oncotarget ; 5(2): 561-74, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24504474

RESUMEN

Hypoxia has emerged as one of the most important drivers of tumor aggression, metastasis, and poor clinical outcome in many cancers.In prostate cancer (PCa), hypoxia has been strongly correlated to biochemical failure and local recurrence. However, current PCa treatment options do not address hypoxic cells highlighting a critical gap in existing therapies and the need for development of therapies that target hypoxic prostate tumor cells. Mammalian orthoreovirus (MRV) is an oncolytic virus that targets tumor cells over normal cells which has been shown to be safe and effective against many cancers in vitro, in animal models, and in human clinical trials. We found that MRVinfects and replicates in hypoxic prostate tumor cells to levels comparable to normoxic cells leading to apoptosis and cell death. In addition, the regulatory subunit (HIF-1α) of the master transcriptional regulator of hypoxia, HIF-1, was significantly downregulated in infected cells. HIF-1α downregulation was found to occur via ubiquitin-dependent proteasome-mediated degradation and translational inhibition. Virus-mediated HIF-1α degradation required the HIF-1α PAS domain and expression of the receptor for activated kinase C (RACK1) protein. These data provide evidence that MRV may be a viable therapeutic option for targeting hypoxic cells and HIF-1α in PCa.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/virología , Infecciones por Reoviridae/metabolismo , Factores de Transcripción/genética , Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Progresión de la Enfermedad , Regulación hacia Abajo , Humanos , Masculino , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/virología , Orthoreovirus de los Mamíferos , Neoplasias de la Próstata/patología , Infecciones por Reoviridae/patología , Transfección
7.
Virology ; 448: 133-45, 2014 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-24314644

RESUMEN

At early times in Mammalian Orthoreovirus (MRV) infection, cytoplasmic inclusions termed stress granules (SGs) are formed as a component of the innate immune response, however, at later times they are no longer present despite continued immune signaling. To investigate the roles of MRV proteins in SG modulation we examined non-structural protein µNS localization relative to SGs in infected and transfected cells. Using a series of mutant plasmids, we mapped the necessary µNS residues for SG localization to amino acids 78 and 79. We examined the capacity of a µNS(78-79) mutant to associate with known viral protein binding partners of µNS and found that it loses association with viral core protein λ2. Finally, we show that while this mutant cannot support de novo viral replication, it is able to rescue replication following siRNA knockdown of µNS. These data suggest that µNS association with SGs, λ2, or both play roles in MRV replication.


Asunto(s)
Gránulos Citoplasmáticos/virología , Orthoreovirus Mamífero 3/metabolismo , Infecciones por Reoviridae/virología , Proteínas del Núcleo Viral/metabolismo , Proteínas no Estructurales Virales/administración & dosificación , Proteínas no Estructurales Virales/química , Replicación Viral , Secuencias de Aminoácidos , Animales , Línea Celular , Humanos , Orthoreovirus Mamífero 3/química , Orthoreovirus Mamífero 3/genética , Unión Proteica , Proteínas del Núcleo Viral/genética , Proteínas no Estructurales Virales/genética
8.
Virology ; 399(1): 153-166, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20097399

RESUMEN

Flaviviruses have a positive-sense, single-stranded RNA genome of approximately 11 kb, encoding a large polyprotein that is cleaved to produce approximately 10 mature proteins. Cell fusing agent virus, Kamiti River virus, Culex flavivirus and several recently discovered flaviviruses have no known vertebrate host and apparently infect only insects. We present compelling bioinformatic evidence for a 253-295 codon overlapping gene (designated fifo) conserved throughout these insect-specific flaviviruses and immunofluorescent detection of its product. Fifo overlaps the NS2A/NS2B coding sequence in the -1/+2 reading frame and is most likely expressed as a trans-frame fusion protein via ribosomal frameshifting at a conserved GGAUUUY slippery heptanucleotide with 3'-adjacent RNA secondary structure (which stimulates efficient frameshifting in vitro). The discovery bears striking parallels to the recently discovered ribosomal frameshifting site in the NS2A coding sequence of the Japanese encephalitis serogroup of flaviviruses and suggests that programmed ribosomal frameshifting may be more widespread in flaviviruses than currently realized.


Asunto(s)
Flavivirus/genética , Sistema de Lectura Ribosómico/genética , Genes Sobrepuestos/genética , Aedes/virología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Codón/genética , Secuencia Conservada/genética , Culex/virología , Genoma Viral/genética , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta/genética , Filogenia , Alineación de Secuencia , Proteínas Virales/genética
9.
J Virol ; 84(2): 867-82, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19889754

RESUMEN

Virally induced structures called viral factories form throughout the cytoplasm of cells infected with mammalian orthoreoviruses (MRV). When expressed alone in cells, MRV nonstructural protein microNS forms factory-like structures very similar in appearance to viral factories, suggesting that it is involved in forming the structural matrix of these structures. microNS also associates with MRV core particles; the core proteins mu2, lambda1, lambda2, lambda3, and sigma2; and the RNA-binding nonstructural protein sigmaNS. These multiple associations result in the recruitment or retention of these viral proteins or particles at factory-like structures. In this study, we identified the regions of microNS necessary and sufficient for these associations and additionally examined the localization of viral RNA synthesis in infected cells. We found that short regions within the amino-terminal 220 residues of microNS are necessary for associations with core particles and necessary and sufficient for associations with the proteins mu2, lambda1, lambda2, sigma2, and sigmaNS. We also found that only the lambda3 protein associates with the carboxyl-terminal one-third of microNS and that viral RNA is synthesized within viral factories. These results suggest that microNS may act as a cytoplasmic scaffolding protein involved in localizing and coordinating viral replication or assembly intermediates for the efficient production of progeny core particles during MRV infection.


Asunto(s)
Citoplasma/ultraestructura , Cuerpos de Inclusión/metabolismo , Orthoreovirus de los Mamíferos/patogenicidad , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/ultraestructura , Orthoreovirus de los Mamíferos/genética , Orthoreovirus de los Mamíferos/metabolismo , ARN Viral/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Proteínas no Estructurales Virales/genética , Ensamble de Virus
10.
Mol Cell Proteomics ; 6(6): 1027-38, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17339631

RESUMEN

Protein-protein associations are vital to cellular functions. Here we describe a helpful new method to demonstrate protein-protein associations inside cells based on the capacity of orthoreovirus protein muNS to form large cytoplasmic inclusions, easily visualized by light microscopy, and to recruit other proteins to these structures in a specific manner. We introduce this technology by the identification of a sixth orthoreovirus protein, RNA-dependent RNA polymerase lambda3, that was recruited to the structures through an association with muNS. We then established the broader utility of this technology by using a truncated, fluorescently tagged form of muNS as a fusion platform to present the mammalian tumor suppressor p53, which strongly recruited its known interactor simian virus 40 large T antigen to the muNS-derived structures. In both examples, we further localized a region of the recruited protein that is key to its recruitment. Using either endogenous p53 or a second fluorescently tagged fusion of p53 with the rotavirus NSP5 protein, we demonstrated p53 oligomerization as well as p53 association with another of its cellular interaction partners, the CREB-binding proteins, within the inclusions. Furthermore using the p53-fused fluorescent muNS platform in conjunction with three-color microscopy, we identified a ternary complex comprising p53, simian virus 40 large T antigen, and retinoblastoma protein. The new method is technically simple, uses commonly available resources, and is adaptable to high throughput formats.


Asunto(s)
Orthoreovirus/química , Proteínas Virales/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Células COS , Chlorocebus aethiops , Estructuras Citoplasmáticas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo
11.
J Virol ; 78(19): 10291-302, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15367595

RESUMEN

Reovirus replication and assembly are thought to occur within cytoplasmic inclusion bodies, which we call viral factories. A strain-dependent difference in the morphology of these structures reflects more effective microtubule association by the mu2 core proteins of some viral strains, which form filamentous factories, than by those of others, which form globular factories. For this report, we identified and characterized another strain-dependent attribute of the factories, namely, the extent to which they colocalized with conjugated ubiquitin (cUb). Among 16 laboratory strains and field isolates, the extent of factory costaining for cUb paralleled factory morphology, with globular strains exhibiting higher levels by far. In reassortant viruses, factory costaining for cUb mapped primarily to the mu2-encoding M1 genome segment, although contributions by the lambda3- and lambda2-encoding L1 and L2 genome segments were also evident. Immunoprecipitations revealed that cells infected with globular strains contained higher levels of ubiquitinated mu2 (Ub-mu2). In M1-transfected cells, cUb commonly colocalized with aggregates formed by mu2 from globular strains but not with microtubules coated by mu2 from filamentous strains, and immunoprecipitations revealed that mu2 from globular strains displayed higher levels of Ub-mu2. Allelic changes at mu2 residue 208 determined these differences. Nocodazole treatment of cells infected with filamentous strains resulted in globular factories that still showed low levels of costaining for cUb, indicating that higher levels of costaining were not a direct result of decreased microtubule association. The factories of globular strains, or their mu2 proteins expressed in transfected cells, were furthermore shown to gain microtubule association and to lose colocalization with cUb when cells were grown at reduced temperature. From the sum of these findings, we propose that mu2 from globular strains is more prone to temperature-dependent misfolding and as a result displays increased aggregation, increased levels of Ub-mu2, and decreased association with microtubules. Because so few of the viral strains formed factories that were regularly associated with ubiquitinated proteins, we conclude that reovirus factories are generally distinct from cellular aggresomes.


Asunto(s)
Cuerpos de Inclusión Viral/ultraestructura , Orthoreovirus/crecimiento & desarrollo , Orthoreovirus/genética , Ubiquitina/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Genes Virales , Cuerpos de Inclusión Viral/metabolismo , Microtúbulos/química , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Orthoreovirus/metabolismo , Fenotipo , Pliegue de Proteína , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Reordenados/genética , Virus Reordenados/crecimiento & desarrollo , Virus Reordenados/metabolismo , Temperatura , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Replicación Viral/genética
12.
J Virol ; 78(19): 10695-705, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15367636

RESUMEN

Nonfusogenic mammalian orthoreovirus (reovirus) is an enteric pathogen of mice and a useful model for studies of how an enteric virus crosses the mucosal barrier of its host and is subject to control by the mucosal immune system. We recently generated and characterized a new murine immunoglobulin A (IgA)-class monoclonal antibody (MAb), 1E1, that binds to the adhesin fiber, sigma1, of reovirus type 1 Lang (T1L) and thereby neutralizes the infectivity of that strain in cell culture. 1E1 is produced in hybridoma cultures as a mixture of monomers, dimers, and higher polymers and is protective against peroral challenges with T1L either when the MAb is passively administered or when it is secreted into the intestines of mice bearing subcutaneous hybridoma tumors. In the present study, selection and analysis of mutants resistant to neutralization by 1E1 identified the region of T1L sigma1 to which the MAb binds. The region bound by a previously characterized type 1 sigma1-specific neutralizing IgG MAb, 5C6, was identified in the same way. Each of the 15 mutants isolated and analyzed was found to be much less sensitive to neutralization by either 1E1 or 5C6, suggesting the two MAbs bind to largely overlapping regions of sigma1. The tested mutants retained the capacity to recognize specific glycoconjugate receptors on rabbit M cells and cultured epithelial cells, even though viral binding to epithelial cells was inhibited by both MAbs. S1 sequence determinations for 12 of the mutants identified sigma1 mutations at four positions between residues 415 and 447, which contribute to forming the receptor-binding head domain. When aligned with the sigma1 sequence of reovirus type 3 Dearing (T3D) and mapped onto the previously reported crystal structure of the T3D sigma1 trimer, the four positions cluster on the side of the sigma1 head, across the interface between two subunits. Three such interface-spanning epitopes are thus present per sigma1 trimer and require the intact quaternary structure of the head domain for MAb binding. Identification of these intersubunit epitopes on sigma1 opens the way for further studies of the mechanisms of antibody-based neutralization and protection with type 1 reoviruses.


Asunto(s)
Proteínas de la Cápside/inmunología , Mapeo Epitopo , Epítopos/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Orthoreovirus de los Mamíferos/inmunología , Sustitución de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Modelos Moleculares , Mutación Missense , Pruebas de Neutralización , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Subunidades de Proteína/inmunología , Receptores Virales , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/virología , Ensamble de Virus
13.
J Virol ; 78(4): 1882-92, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747553

RESUMEN

Mammalian reoviruses are thought to assemble and replicate within cytoplasmic, nonmembranous structures called viral factories. The viral nonstructural protein mu NS forms factory-like globular inclusions when expressed in the absence of other viral proteins and binds to the surfaces of the viral core particles in vitro. Given these previous observations, we hypothesized that one or more of the core surface proteins may be recruited to viral factories through specific associations with mu NS. We found that all three of these proteins--lambda 1, lambda 2, and sigma 2--localized to factories in infected cells but were diffusely distributed through the cytoplasm and nucleus when each was separately expressed in the absence of other viral proteins. When separately coexpressed with mu NS, on the other hand, each core surface protein colocalized with mu NS in globular inclusions, supporting the initial hypothesis. We also found that lambda 1, lambda 2, and sigma 2 each localized to filamentous inclusions formed upon the coexpression of mu NS and mu 2, a structurally minor core protein that associates with microtubules. The first 40 residues of mu NS, which are required for association with mu 2 and the RNA-binding nonstructural protein sigma NS, were not required for association with any of the three core surface proteins. When coexpressed with mu 2 in the absence of mu NS, each of the core surface proteins was diffusely distributed and displayed only sporadic, weak associations with mu 2 on filaments. Many of the core particles that entered the cytoplasm of cycloheximide-treated cells following entry and partial uncoating were recruited to inclusions of mu NS that had been preformed in those cells, providing evidence that mu NS can bind to the surfaces of cores in vivo. These findings expand a model for how viral and cellular components are recruited to the viral factories in infected cells and provide further evidence for the central but distinct roles of viral proteins mu NS and mu 2 in this process.


Asunto(s)
Cuerpos de Inclusión Viral/metabolismo , Reoviridae/metabolismo , Proteínas del Núcleo Viral/metabolismo , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus , Animales , Proteínas de la Cápside , Línea Celular , Proteínas de Unión al ADN , Humanos , Ratones , Microscopía Fluorescente , Proteínas de Unión al ARN , Proteínas Virales/metabolismo , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA