Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(45): 11603-11607, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30337483

RESUMEN

Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that causes immunosuppression, paralysis, and deadly lymphomas in chickens. In infected animals, B cells are efficiently infected and are thought to amplify the virus and transfer it to T cells. MDV subsequently establishes latency in T cells and transforms CD4+ T cells, resulting in fatal lymphomas. Despite many years of research, the exact role of the different B and T cell subsets in MDV pathogenesis remains poorly understood, mostly due to the lack of reverse genetics in chickens. Recently, Ig heavy chain J gene segment knockout (JH-KO) chickens lacking mature and peripheral B cells have been generated. To determine the role of these B cells in MDV pathogenesis, we infected JH-KO chickens with the very virulent MDV RB1B strain. Surprisingly, viral load in the blood of infected animals was not altered in the absence of B cells. More importantly, disease and tumor incidence in JH-KO chickens was comparable to wild-type animals, suggesting that both mature and peripheral B cells are dispensable for MDV pathogenesis. Intriguingly, MDV efficiently replicated in the bursa of Fabricius in JH-KO animals, while spread of the virus to the spleen and thymus was delayed. In the absence of B cells, MDV readily infected CD4+ and CD8+ T cells, allowing efficient virus replication in the lymphoid organs and transformation of T cells. Taken together, our data change the dogma of the central role of B cells, and thereby provide important insights into MDV pathogenesis.


Asunto(s)
Linfocitos B/inmunología , Genoma Viral , Herpesvirus Gallináceo 2/patogenicidad , Linfoma/patología , Enfermedad de Marek/patología , Virus Oncogénicos/patogenicidad , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Bolsa de Fabricio/inmunología , Bolsa de Fabricio/virología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Embrión de Pollo , Pollos , ADN Viral/genética , ADN Viral/inmunología , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 2/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Recuento de Linfocitos , Linfoma/genética , Linfoma/inmunología , Linfoma/virología , Enfermedad de Marek/genética , Enfermedad de Marek/inmunología , Enfermedad de Marek/virología , Virus Oncogénicos/genética , Virus Oncogénicos/inmunología , Bazo/inmunología , Bazo/virología , Timo/inmunología , Timo/virología , Carga Viral , Virulencia , Replicación Viral
2.
J Med Virol ; 91(11): 1960-1969, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31317546

RESUMEN

Hepatitis E is an important global disease, causing outbreaks of acute hepatitis in many developing countries and sporadic cases in industrialized countries. Hepatitis E virus (HEV) infection typically causes self-limiting acute hepatitis but can also progress to chronic disease in immunocompromised individuals. The immune response necessary for the prevention of chronic infection is T cell-dependent; however, the arm of cellular immunity responsible for this protection is not currently known. To investigate the contribution of humoral immunity in control of HEV infection and prevention of chronicity, we experimentally infected 20 wild-type (WT) and 18 immunoglobulin knockout (JH-KO) chickens with a chicken strain of HEV (avian HEV). Four weeks postinfection (wpi) with avian HEV, JH-KO chickens were unable to elicit anti-HEV antibody but had statistically significantly lower liver lesion scores than the WT chickens. At 16 wpi, viral RNA in fecal material and liver, and severe liver lesions were undetectable in both groups. To determine the role of cytotoxic lymphocytes in the prevention of chronicity, we infected 20 WT and 20 cyclosporine and CD8+ antibody-treated chickens with the same strain of avian HEV. The CD8 + lymphocyte-depleted, HEV-infected chickens had higher incidences of prolonged fecal viral shedding and statistically significantly higher liver lesion scores than the untreated, HEV-infected birds at 16 wpi. The results indicate that CD8 + lymphocytes are required for viral clearance and reduction of liver lesions in HEV infection while antibodies are not necessary for viral clearance but may contribute to the development of liver lesions in acute HEV infection.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Anticuerpos Antihepatitis/sangre , Hepatitis Viral Animal/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Infecciones por Virus ARN/veterinaria , Animales , Pollos/inmunología , Heces/virología , Técnicas de Inactivación de Genes , Hepatitis Viral Animal/inmunología , Hepevirus , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulinas/genética , Hígado/patología , Hígado/virología , Depleción Linfocítica , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/prevención & control , ARN Viral/análisis , Esparcimiento de Virus
3.
Eur J Immunol ; 46(9): 2137-48, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27392810

RESUMEN

Since the discovery of antibody-producing B cells in chickens six decades ago, chickens have been a model for B-cell development in gut-associated lymphoid tissue species. Here we describe targeting of the immunoglobulin light chain locus by homologous recombination in chicken primordial germ cells (PGCs) and generation of VJCL knockout chickens. In contrast to immunoglobulin heavy chain knockout chickens, which completely lack mature B cells, homozygous light chain knockout (IgL(-/-) ) chickens have a small population of B lineage cells that develop in the bursa and migrate to the periphery. This population of B cells expresses the immunoglobulin heavy chain molecule on the cell surface. Soluble heavy-chain-only IgM and IgY proteins of reduced molecular weight were detectable in plasma in 4-week-old IgL(-/-) chickens, and antigen-specific IgM and IgY heavy chain proteins were produced in response to immunization. Circulating heavy-chain-only IgM showed a deletion of the CH1 domain of the constant region enabling the immunoglobulin heavy chain to be secreted in the absence of the light chain. Our data suggest that the heavy chain by itself is enough to support all the important steps in B-cell development in a gut-associated lymphoid tissue species.


Asunto(s)
Anticuerpos/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Expresión Génica , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Animales , Animales Modificados Genéticamente , Anticuerpos/inmunología , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Pollos , Eliminación de Gen , Técnicas de Inactivación de Genes , Orden Génico , Marcación de Gen , Vectores Genéticos/genética , Cadenas Ligeras de Inmunoglobulina/química , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Dominios Proteicos/genética
4.
Proc Natl Acad Sci U S A ; 110(50): 20170-5, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24282302

RESUMEN

Gene targeting by homologous recombination or by sequence-specific nucleases allows the precise modification of genomes and genes to elucidate their functions. Although gene targeting has been used extensively to modify the genomes of mammals, fish, and amphibians, a targeting technology has not been available for the avian genome. Many of the principles of humoral immunity were discovered in chickens, yet the lack of gene targeting technologies in birds has limited biomedical research using this species. Here we describe targeting the joining (J) gene segment of the chicken Ig heavy chain gene by homologous recombination in primordial germ cells to establish fully transgenic chickens carrying the knockout. In homozygous knockouts, Ig heavy chain production is eliminated, and no antibody response is elicited on immunization. Migration of B-lineage precursors into the bursa of Fabricius is unaffected, whereas development into mature B cells and migration from the bursa are blocked in the mutants. Other cell types in the immune system appear normal. Chickens lacking the peripheral B-cell population will provide a unique experimental model to study avian immune responses to infectious disease. More generally, gene targeting in avian primordial germ cells will foster advances in diverse fields of biomedical research such as virology, stem cells, and developmental biology, and provide unique approaches in biotechnology, particularly in the field of antibody discovery.


Asunto(s)
Linfocitos B/citología , Pollos/genética , Técnicas de Inactivación de Genes/métodos , Ingeniería Genética/métodos , Células Germinativas/química , Cadenas Pesadas de Inmunoglobulina/genética , Animales , Linfocitos B/metabolismo , Southern Blotting , Pollos/inmunología , Metilación de ADN , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Genotipo , Células Germinativas/metabolismo , Inmunohistoquímica
5.
Poult Sci ; 94(4): 799-803, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25828572

RESUMEN

During the past decade, modifications to the chicken genome have evolved from random insertions of small transgenes using viral vectors to site-specific deletions using homologous recombination vectors and nontargeted insertions of large transgenes using phi-31 integrase. Primordial germ cells (PGC) and gonocytes are the germline-competent cell lines in which targeted modifications and large transgenes are inserted into the genome. After extended periods of in vitro culture, PGC retain their capacity to form functional gametes when reintroduced in vivo. Rates of stable germline modification vary from 1×10(-5) for nontargeted insertions to 1×10(-8) for targeted insertions. Following transfection, clonally derived cell lines are expanded, injected into Stage 13-15 Hamburger and Hamilton embryos, and putative chimeras are incubated to term in surrogate shells. Green fluorescent protein (GFP) is incorporated into transgenes to reveal the presence of genetically modified PGC in culture and the extent of colonization of the gonad during the first week posthatch. If the extent of colonization is adequate, cohorts of putative chimeras are reared to sexual maturity. Semen is collected and the contribution from donor PGC is estimated by evaluating GFP expression using flow cytometry and PCR. The most promising candidates are selected for breeding to obtain G1 heterozygote offspring. To date, this protocol has been used to (1) knockout the immunoglobulin heavy and light chain genes and produce chickens lacking humoral immunity, (2) insert human V genes and arrays of pseudo V genes into the heavy and light immunoglobulin loci to produce chickens making antibodies with human V regions, (3) insert GFP into nontargeted locations within the genome to produce chickens expressing GFP, and (4) insert Cre recombinase into the genome to produce chickens that excise sequences of DNA flanked by loxP sites.


Asunto(s)
Pollos/genética , Genoma , Mutagénesis Insercional , Transgenes , Animales , Humanos
6.
Biol Reprod ; 90(1): 15, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24337317

RESUMEN

Avian cell lines derived from germinal crescent primordial germ cells and gonadal gonocytes with long-term proliferative capacity in vitro and their subsequent rates of colonization and germline transmission are described. In general, male cultures proliferate more rapidly than female cultures although both can be developed into cell lines of >2 × 10(6) cells, at which time, they can be grown indefinitely and a cell bank can be established. All the cell lines injected into embryos transmitted through the germline with the percentage of germline transmission of both male and female cell lines varying from single digits to the high 90s. The derivation of these primordial germ cell and gonadal cell lines and the subsequent robustness of germline transmission validates these cells as suitable for establishment of lines of chickens bearing novel genetic modifications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Germinativas/citología , Gónadas/citología , Animales , Animales Modificados Genéticamente , Forma de la Célula , Células Cultivadas , Embrión de Pollo , Quimera/embriología , Células Madre Embrionarias/citología , Femenino , Masculino , Factores de Tiempo
7.
Nature ; 441(7094): 766-9, 2006 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-16760981

RESUMEN

Primordial germ cells (PGCs) are the precursors of sperm and eggs. In most animals, segregation of the germ line from the somatic lineages is one of the earliest events in development; in avian embryos, PGCs are first identified in an extra-embryonic region, the germinal crescent, after approximately 18 h of incubation. After 50-55 h of development, PGCs migrate to the gonad and subsequently produce functional sperm and oocytes. So far, cultures of PGCs that remain restricted to the germ line have not been reported in any species. Here we show that chicken PGCs can be isolated, cultured and genetically modified while maintaining their commitment to the germ line. Furthermore, we show that chicken PGCs can be induced in vitro to differentiate into embryonic germ cells that contribute to somatic tissues. Retention of the commitment of PGCs to the germ line after extended periods in culture and after genetic modification combined with their capacity to acquire somatic competence in vitro provides a new model for developmental biology. The utility of the model is enhanced by the accessibility of the avian embryo, which facilitates access to the earliest stages of development and supplies a facile route for the reintroduction of PGCs into the embryonic vasculature. In addition, these attributes create new opportunities to manipulate the genome of chickens for agricultural and pharmaceutical applications.


Asunto(s)
Linaje de la Célula , Pollos/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Mutación de Línea Germinal/genética , Células Madre/citología , Células Madre/metabolismo , Animales , Línea Celular , Células Cultivadas , Embrión de Pollo , Femenino , Citometría de Flujo , Ingeniería Genética/métodos , Genoma/genética , Células Germinativas/trasplante , Cariotipificación , Masculino , Óvulo/citología , Óvulo/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo , Trasplante de Células Madre
8.
Mol Reprod Dev ; 75(7): 1163-75, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18213680

RESUMEN

The genome of germline committed cells is thought to be protected by mechanisms of transcriptional silencing, posing a barrier to transgenesis using cultured germline cells. We found that selection for transgene integration into the primordial germ cell genome required that the transgenes be flanked by the chicken beta-globin insulator. However, integration frequency was low, and sequencing of the insertion sites revealed that the transgenes preferentially inserted into active promoter regions, implying that silencing prohibited recovery of insertions in other regions. Much higher frequencies of integration were achieved when the phiC31 integrase was used to insert transgenes into endogenous pseudo attP sites. Despite the evidence for transcriptional silencing in PGCs, gene targeting of a nonexpressed gene was also achieved. The ability to make genetic modifications in PGCs provides unprecedented opportunities to study the biology of PGCs, as well as produce transgenic chickens for applications in biotechnology and developmental biology.


Asunto(s)
Marcación de Gen/métodos , Células Germinativas/fisiología , Integrasas/genética , Animales , Secuencia de Bases , Embrión de Pollo/fisiología , Mapeo Cromosómico , Clonación Molecular , Cartilla de ADN , ADN Circular/genética , Células Germinativas/enzimología , Datos de Secuencia Molecular
9.
Nat Biotechnol ; 23(9): 1159-69, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16127450

RESUMEN

The tubular gland of the chicken oviduct is an attractive system for protein expression as large quantities of proteins are deposited in the egg, the production of eggs is easily scalable and good manufacturing practices for therapeutics from eggs have been established. Here we examined the ability of upstream and downstream DNA sequences of ovalbumin, a protein produced exclusively in very high quantities in chicken egg white, to drive tissue-specific expression of human mAb in chicken eggs. To accommodate these large regulatory regions, we established and transfected lines of chicken embryonic stem (cES) cells and formed chimeras that express mAb from cES cell-derived tubular gland cells. Eggs from high-grade chimeras contained up to 3 mg of mAb that possesses enhanced antibody-dependent cellular cytotoxicity (ADCC), nonantigenic glycosylation, acceptable half-life, excellent antigen recognition and good rates of internalization.


Asunto(s)
Anticuerpos Monoclonales/química , Animales , Southern Blotting , Western Blotting , Células CHO , Rastreo Diferencial de Calorimetría , Carbohidratos/química , Pollos , Cricetinae , ADN/metabolismo , Clara de Huevo , Embrión de Mamíferos/citología , Embrión no Mamífero , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos , Genoma , Glicosilación , Humanos , Inmunoglobulina G , Inmunohistoquímica , Focalización Isoeléctrica , Ratones , Ratones Endogámicos BALB C , Modelos Genéticos , Monosacáridos/química , Oligosacáridos/química , Ovalbúmina/genética , Ovalbúmina/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Ionización de Electrospray , Células Madre/citología
10.
MAbs ; 10(1): 71-80, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29035625

RESUMEN

Transgenic animal platforms for the discovery of human monoclonal antibodies have been developed in mice, rats, rabbits and cows. The immune response to human proteins is limited in these animals by their tolerance to mammalian-conserved epitopes. To expand the range of epitopes that are accessible, we have chosen an animal host that is less phylogenetically related to humans. Specifically, we generated transgenic chickens expressing antibodies from immunoglobulin heavy and light chain loci containing human variable regions and chicken constant regions. From these birds, paired human light and heavy chain variable regions are recovered and cloned as fully human recombinant antibodies. The human antibody-expressing chickens exhibit normal B cell development and raise immune responses to conserved human proteins that are not immunogenic in mice. Fully human monoclonal antibodies can be recovered with sub-nanomolar affinities. Binning data of antibodies to a human protein show epitope coverage similar to wild type chickens, which we previously showed is broader than that produced from rodent immunizations.


Asunto(s)
Anticuerpos Monoclonales Humanizados/biosíntesis , Anticuerpos Monoclonales Humanizados/inmunología , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Antígenos/inmunología , Pollos/inmunología , Epítopos/inmunología , Inmunoglobulinas/inmunología , Animales , Animales Modificados Genéticamente , Antígenos/administración & dosificación , Linfocitos B/inmunología , Pollos/sangre , Pollos/genética , Mapeo Epitopo , Humanos , Inmunización , Inmunoglobulinas/sangre , Inmunoglobulinas/genética , Especificidad de la Especie , Linfocitos T/inmunología
11.
Mech Dev ; 123(1): 31-41, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16325380

RESUMEN

Male and female embryonic stem (ES) cell lines were derived from the area pellucidae of Stage X (EG&K) chicken embryos. These ES cell lines were grown in culture for extended periods of time and the majority of the cells retained a diploid karyotype. When reintroduced into Stage VI-X (EG&K) recipient embryos, the cES cells were able to contribute to all somatic tissues. By combining irradiation of the recipient embryo with exposure of the cES cells to the embryonic environment in diapause, a high frequency and extent of chimerism was obtained. High-grade chimeras, indistinguishable from the donor phenotype by feather pigmentation, were produced. A transgene encoding GFP was incorporated into the genome of cES cells under control of the ubiquitous promoter CX and GFP was widely expressed in somatic tissues. Although cES cells made extensive contributions to the somatic tissues, contribution to the germline was not observed.


Asunto(s)
Embrión de Pollo/citología , Quimera , Células Madre Pluripotentes/citología , Animales , Animales Modificados Genéticamente , Proteínas Aviares/genética , Secuencia de Bases , Línea Celular , Proliferación Celular , Embrión de Pollo/metabolismo , Pollos , Quimera/genética , ADN Complementario/genética , Diploidia , Trastornos del Desarrollo Sexual , Femenino , Células Germinativas , Masculino , Proteínas del Tejido Nervioso/genética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Trasplante de Células Madre
12.
PLoS One ; 8(11): e80108, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278246

RESUMEN

Transgenic chickens expressing human sequence antibodies would be a powerful tool to access human targets and epitopes that have been intractable in mammalian hosts because of tolerance to conserved proteins. To foster the development of the chicken platform, it is beneficial to validate transgene constructs using a rapid, cell culture-based method prior to generating fully transgenic birds. We describe a method for the expression of human immunoglobulin variable regions in the chicken DT40 B cell line and the further diversification of these genes by gene conversion. Chicken VL and VH loci were knocked out in DT40 cells and replaced with human VK and VH genes. To achieve gene conversion of human genes in chicken B cells, synthetic human pseudogene arrays were inserted upstream of the functional human VK and VH regions. Proper expression of chimeric IgM comprised of human variable regions and chicken constant regions is shown. Most importantly, sequencing of DT40 genetic variants confirmed that the human pseudogene arrays contributed to the generation of diversity through gene conversion at both the Igl and Igh loci. These data show that engineered pseudogene arrays produce a diverse pool of human antibody sequences in chicken B cells, and suggest that these constructs will express a functional repertoire of chimeric antibodies in transgenic chickens.


Asunto(s)
Anticuerpos/genética , Linfocitos B/metabolismo , Pollos/genética , Conversión Génica , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Anticuerpos/química , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Citometría de Flujo , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
13.
PLoS One ; 7(5): e35664, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22629301

RESUMEN

In birds, the primordial germ cell (PGC) lineage separates from the soma within 24 h following fertilization. Here we show that the endogenous population of about 200 PGCs from a single chicken embryo can be expanded one million fold in culture. When cultured PGCs are injected into a xenogeneic embryo at an equivalent stage of development, they colonize the testis. At sexual maturity, these donor PGCs undergo spermatogenesis in the xenogeneic host and become functional sperm. Insemination of semen from the xenogeneic host into females from the donor species produces normal offspring from the donor species. In our model system, the donor species is chicken (Gallus domesticus) and the recipient species is guinea fowl (Numida meleagris), a member of a different avian family, suggesting that the mechanisms controlling proliferation of the germline are highly conserved within birds. From a pragmatic perspective, these data are the basis of a novel strategy to produce endangered species of birds using domesticated hosts that are both tractable and fecund.


Asunto(s)
Quimera/genética , Galliformes/genética , Células Germinativas/citología , Animales , Línea Celular , Células Cultivadas , Femenino , Masculino
15.
Transgenic Res ; 15(5): 521-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16953329

RESUMEN

After 25 years, the search for the avian cell that can be cultured indefinitely, genetically modified, and clonally derived while retaining its ability to enter the germline has ended. van de Lavoir et al. [2006a, Nature 441:766-769] have defined the conditions for culture and genetic modification of primordial germ cells (PGCs) and shown that these cells are transmitted at high rates through the germline. The advent of this technology provides the ability to introduce transgenes of any size and to make site-specific changes to the genome. Although PGCs are committed to the germline, they can be induced into somatically committed embryonic germ (EG) cells by changing the culture conditions. EG cells resemble embryonic stem (ES) cells that are also committed to the somatic lineages (van de Lavoir 2006b, Mech Dev 123:31-41). These cell-based systems facilitate insertion of larger transgenes that provide high level, developmentally regulated and tissue-specific expression in transgenic chimeras and their offspring. Following introduction of a transgene, high-grade somatic chimeras can be made with ES and EG cells within 4 weeks and 4 months respectively, allowing quick assessment of the transgenic phenotype. Following introduction of a tansgene into PGCs, high-grade germline chimeras can be made within 8-9 weeks and the high rate of germline transmission of G0 chimeras produces a large cohort of transgenic chicks in 16-17 weeks. PGC, EG and ES cells can be grown in conventional laboratory settings and small flocks of recipient birds or third-party vendors can supply recipient embryos to make somatic and/or germline chimeras. In general, animal management is routine although some specialized equipment and technical skill is required to incubate chimeras in surrogate shells.


Asunto(s)
Animales Modificados Genéticamente , Aves/genética , Técnicas de Transferencia de Gen , Animales , Células Madre Embrionarias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA