Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 14(12): e0225021, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31821343

RESUMEN

Type 1, or autoimmune, diabetes is caused by the T-cell mediated destruction of the insulin-producing pancreatic beta cells. Non-obese diabetic (NOD) mice spontaneously develop autoimmune diabetes akin to human type 1 diabetes. For this reason, the NOD mouse has been the preeminent murine model for human type 1 diabetes research for several decades. However, humanized mouse models are highly sought after because they offer both the experimental tractability of a mouse model and the clinical relevance of human-based research. Autoimmune T-cell responses against insulin, and its precursor proinsulin, play central roles in the autoimmune responses against pancreatic beta cells in both humans and NOD mice. As a first step towards developing a murine model of the human autoimmune response against pancreatic beta cells we set out to replace the murine insulin 1 gene (Ins1) with the human insulin gene (Ins) using CRISPR/Cas9. Here we describe a NOD mouse strain that expresses human insulin in place of murine insulin 1, referred to as HuPI. HuPI mice express human insulin, and C-peptide, in their serum and pancreata and have normal glucose tolerance. Compared with wild type NOD mice, the incidence of diabetes is much lower in HuPI mice. Only 15-20% of HuPI mice developed diabetes after 300 days, compared to more than 60% of unmodified NOD mice. Immune-cell infiltration into the pancreatic islets of HuPI mice was not detectable at 100 days but was clearly evident by 300 days. This work highlights the feasibility of using CRISPR/Cas9 to create mouse models of human diseases that express proteins pivotal to the human disease. Furthermore, it reveals that even subtle changes in proinsulin protect NOD mice from diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Animales , Sistemas CRISPR-Cas , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Páncreas/metabolismo
2.
Diabetologia ; 62(3): 351-356, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30402774

RESUMEN

Type 1 diabetes is an autoimmune disease caused by T cell-mediated destruction of pancreatic insulin-producing beta cells. The epitopes recognised by pathogenic T cells in human type 1 diabetes are poorly defined; however, a growing body of evidence suggests that T cell responses against neoepitopes contribute to beta cell destruction in type 1 diabetes. Neoepitopes are formed when self-proteins undergo post-translational modification to create a new epitope that is recognised by T- or B cells. Here we review the role of human T cell responses against neoepitopes in the immune pathogenesis of type 1 diabetes. Specifically, we review the different approaches to identifying neoepitopes relevant to human type 1 diabetes and outline several advances in this field that have occurred over the past few years. We also discuss the application of neoepitopes to the development of antigen-specific therapies for type 1 diabetes and the unresolved challenges that need to be overcome before the full repertoire of neoepitopes recognised by pathogenic human T cells in type 1 diabetes can be determined. This information may then be used to develop antigen-specific therapies for type 1 diabetes and assays to monitor changes in pathogenic, beta cell-specific T cell responses.


Asunto(s)
Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/inmunología , Epítopos/inmunología , Células Secretoras de Insulina/inmunología , Animales , Autoantígenos/inmunología , Humanos
3.
Proc Natl Acad Sci U S A ; 115(42): 10732-10737, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30275329

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing beta cells, found within the islets of Langerhans in the pancreas, are destroyed by islet-infiltrating T cells. Identifying the antigenic targets of beta-cell reactive T cells is critical to gain insight into the pathogenesis of T1D and develop antigen-specific immunotherapies. Several lines of evidence indicate that insulin is an important target of T cells in T1D. Because many human islet-infiltrating CD4+ T cells recognize C-peptide-derived epitopes, we hypothesized that full-length C-peptide (PI33-63), the peptide excised from proinsulin as it is converted to insulin, is a target of CD4+ T cells in people with T1D. CD4+ T cell responses to full-length C-peptide were detected in the blood of: 14 of 23 (>60%) people with recent-onset T1D, 2 of 15 (>13%) people with long-standing T1D, and 1 of 13 (<8%) HLA-matched people without T1D. C-peptide-specific CD4+ T cell clones, isolated from six people with T1D, recognized epitopes from the entire 31 amino acids of C-peptide. Eighty-six percent (19 of 22) of the C-peptide-specific clones were restricted by HLA-DQ8, HLA-DQ2, HLA-DQ8trans, or HLA-DQ2trans, HLA alleles strongly associated with risk of T1D. We also found that full-length C-peptide was a much more potent agonist of some CD4+ T cell clones than an 18mer peptide encompassing the cognate epitope. Collectively, our findings indicate that proinsulin C-peptide is a key target of autoreactive CD4+ T cells in T1D. Hence, full-length C-peptide is a promising candidate for antigen-specific immunotherapy in T1D.


Asunto(s)
Autoantígenos/inmunología , Péptido C/inmunología , Péptido C/metabolismo , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/diagnóstico , Antígenos HLA/inmunología , Islotes Pancreáticos/inmunología , Proinsulina/inmunología , Adolescente , Adulto , Células Cultivadas , Niño , Preescolar , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Persona de Mediana Edad , Adulto Joven
4.
Immunol Cell Biol ; 96(1): 34-40, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29359347

RESUMEN

For a long time, immunologists have believed that classical CD4+ and CD8+ T cells recognize peptides (referred to as epitopes), derived from protein antigens presented by MHC/HLA class I or II. Over the past 10-15 years, it has become clear that epitopes recognized by CD8+, and more recently CD4+ T cells, can be formed by protein splicing. Here, we review the discovery of spliced epitopes recognized by tumor-specific human CD8+ T cells. We discuss how these epitopes are formed and some of the unusual variants that have been reported. Now, over a decade since the first report, evidence is emerging that spliced CD8+ T-cell epitopes are much more common, and potentially much more important, than previously imagined. Recent work has shown that epitopes recognized by CD4+ T cells can also be formed by protein splicing. We discuss the recent discovery of spliced CD4+ T-cell epitopes and their potential role as targets of autoimmune T-cell responses. Finally, we highlight some of the new questions raised from our growing appreciation of T-cell epitopes formed by peptide splicing.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/metabolismo , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Vacunas de Subunidad/inmunología , Animales , Presentación de Antígeno , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Humanos , Sistema Inmunológico , Inmunomodulación , Activación de Linfocitos
5.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29263105

RESUMEN

Mycoplasmas are bacterial pathogens of a range of animals, including humans, and are a common cause of respiratory disease. However, the host genetic factors that affect resistance to infection or regulate the resulting pulmonary inflammation are not well defined. We and others have previously demonstrated that nonobese diabetic (NOD) mice can be used to investigate disease loci that affect bacterial infection and autoimmune diabetes. Here we show that NOD mice are more susceptible than C57BL/6 (B6) mice to infection with Mycoplasma pulmonis, a natural model of pulmonary mycoplasmosis. The lungs of infected NOD mice had higher loads of M. pulmonis and more severe inflammatory lesions. Moreover, congenic NOD mice that harbored different B6-derived chromosomal intervals enabled identification and localization of a new mycoplasmosis locus, termed Mpr2, on chromosome 13. These congenic NOD mice demonstrated that the B6 allele for Mpr2 reduced the severity of pulmonary inflammation caused by infection with M. pulmonis and that this was associated with altered cytokine and chemokine concentrations in the infected lungs. Mpr2 also colocalizes to the same genomic interval as Listr2 and Idd14, genetic loci linked to listeriosis resistance and autoimmune diabetes susceptibility, respectively, suggesting that allelic variation within these loci may affect the development of both infectious and autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Infecciones por Mycoplasma/genética , Mycoplasma pulmonis/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Femenino , Sitios Genéticos , Humanos , Pulmón/inmunología , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/microbiología , Mycoplasma pulmonis/genética
6.
Mol Immunol ; 82: 19-33, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28006656

RESUMEN

During immune cell activation, serine-derived lipids such as phosphatidylserine and sphingolipids contribute to the formation of protein signaling complexes within the plasma membrane. Altering lipid composition in the cell membrane can subsequently affect immune cell function and the development of autoimmune disease. Serine incorporator 1 (SERINC1) is a putative carrier protein that facilitates synthesis of serine-derived lipids. To determine if SERINC1 has a role in immune cell function and the development of autoimmunity, we characterized a mouse strain in which a retroviral insertion abolishes expression of the Serinc1 transcript. Expression analyses indicated that the Serinc1 transcript is readily detectable and expressed at relatively high levels in wildtype macrophages and lymphocytes. The ablation of Serinc1 expression in these immune cells, however, did not significantly alter serine-derived lipid composition or affect macrophage function and lymphocyte proliferation. Analyses of Serinc1-deficient mice also indicated that systemic ablation of Serinc1 expression did not affect viability, fertility or autoimmune disease susceptibility. These results suggest that Serinc1 is dispensable for certain immune cell functions and does not contribute to previously reported links between lipid composition in immune cells and autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Susceptibilidad a Enfermedades/inmunología , Activación de Linfocitos/inmunología , Activación de Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Animales , Separación Celular , Diabetes Mellitus Experimental/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Metabolismo de los Lípidos/inmunología , Macrófagos/inmunología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Serina/metabolismo
7.
Science ; 351(6274): 711-4, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26912858

RESUMEN

T cell-mediated destruction of insulin-producing ß cells in the pancreas causes type 1 diabetes (T1D). CD4 T cell responses play a central role in ß cell destruction, but the identity of the epitopes recognized by pathogenic CD4 T cells remains unknown. We found that diabetes-inducing CD4 T cell clones isolated from nonobese diabetic mice recognize epitopes formed by covalent cross-linking of proinsulin peptides to other peptides present in ß cell secretory granules. These hybrid insulin peptides (HIPs) are antigenic for CD4 T cells and can be detected by mass spectrometry in ß cells. CD4 T cells from the residual pancreatic islets of two organ donors who had T1D also recognize HIPs. Autoreactive T cells targeting hybrid peptides may explain how immune tolerance is broken in T1D.


Asunto(s)
Péptido C/inmunología , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Epítopos/inmunología , Células Secretoras de Insulina/inmunología , Secuencia de Aminoácidos , Animales , Péptido C/química , Células Clonales , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Tolerancia Inmunológica , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Péptidos/química , Péptidos/inmunología
8.
G3 (Bethesda) ; 5(12): 2903-11, 2015 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-26438296

RESUMEN

A number of different strategies have been used to identify genes for which genetic variation contributes to type 1 diabetes (T1D) pathogenesis. Genetic studies in humans have identified >40 loci that affect the risk for developing T1D, but the underlying causative alleles are often difficult to pinpoint or have subtle biological effects. A complementary strategy to identifying "natural" alleles in the human population is to engineer "artificial" alleles within inbred mouse strains and determine their effect on T1D incidence. We describe the use of the Sleeping Beauty (SB) transposon mutagenesis system in the nonobese diabetic (NOD) mouse strain, which harbors a genetic background predisposed to developing T1D. Mutagenesis in this system is random, but a green fluorescent protein (GFP)-polyA gene trap within the SB transposon enables early detection of mice harboring transposon-disrupted genes. The SB transposon also acts as a molecular tag to, without additional breeding, efficiently identify mutated genes and prioritize mutant mice for further characterization. We show here that the SB transposon is functional in NOD mice and can produce a null allele in a novel candidate gene that increases diabetes incidence. We propose that SB transposon mutagenesis could be used as a complementary strategy to traditional methods to help identify genes that, when disrupted, affect T1D pathogenesis.


Asunto(s)
Elementos Transponibles de ADN , Diabetes Mellitus Tipo 1/genética , Estudios de Asociación Genética , Vectores Genéticos/genética , Mutagénesis Insercional , Animales , Puntos de Rotura del Cromosoma , Modelos Animales de Enfermedad , Femenino , Dosificación de Gen , Expresión Génica , Genes Reporteros , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Mutación , Sitios de Carácter Cuantitativo
9.
Immunogenetics ; 66(7-8): 501-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24906421

RESUMEN

The nonobese diabetic (NOD) mouse strain serves as a genomic standard for assessing how allelic variation for insulin-dependent diabetes (Idd) loci affects the development of autoimmune diabetes. We previously demonstrated that C57BL/6 (B6) mice harbor a more diabetogenic allele than NOD mice for the Idd14 locus when introduced onto the NOD genetic background. New congenic NOD mouse strains, harboring smaller B6-derived intervals on chromosome 13, now localize Idd14 to an ~18-Mb interval and reveal a new locus, Idd31. Notably, the B6 allele for Idd31 confers protection against diabetes, but only in the absence of the diabetogenic B6 allele for Idd14, indicating genetic epistasis between these two loci. Moreover, congenic mice that are more susceptible to diabetes are more resistant to Listeria monocytogenes infection. This result co-localizes Idd14 and Listr2, a resistance locus for listeriosis, to the same genomic interval and indicates that congenic NOD mice may also be useful for localizing resistance loci for infectious disease.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Epistasis Genética/inmunología , Listeriosis/genética , Listeriosis/inmunología , Alelos , Animales , Femenino , Predisposición Genética a la Enfermedad , Fenómenos Inmunogenéticos , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Endogámicos NOD
10.
Dev Biol ; 392(2): 483-93, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24854998

RESUMEN

The vertebrate T-box transcription factor gene Tbx18 performs a vital role in development of multiple organ systems. Tbx18 insufficiency manifests as recessive phenotypes in the upper urinary system, cardiac venous pole, inner ear, and axial skeleton; homozygous null mutant animals die perinatally. Here, we report a new regulatory mutation of Tbx18, a reciprocal translocation breaking 78kbp downstream of the gene. 12Gso homozygotes present urinary and vertebral defects very similar to those associated with Tbx18-null mutations, but 12Gso is clearly not a global null allele since homozygotes survive into adulthood. We show that 12Gso down-regulates Tbx18 expression in a manner that is both spatially- and temporally-specific; combined with other data, the mutation points particularly to the presence of an essential urogenital enhancer located near the translocation breakpoint site. In support of this hypothesis, we identify a distal enhancer element, ECR1, which is active in developing urogenital and other tissues; we propose that disruption of this element leads to premature loss of Tbx18 function in 12Gso mutant mice. These data reveal a long-range regulatory architecture extending far downstream of Tbx18, identify a novel and likely essential urogenital enhancer, and introduce a new tool for dissecting postnatal phenotypes associated with dysregulation of Tbx18.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/genética , Proteínas de Dominio T Box/metabolismo , Sistema Urogenital/embriología , Azul Alcián , Animales , Antraquinonas , Secuencia de Bases , Mapeo Cromosómico , Cartilla de ADN/genética , Técnicas Histológicas , Inmunohistoquímica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Proteínas de Dominio T Box/genética , Translocación Genética/genética , Sistema Urogenital/metabolismo
11.
Genome Res ; 20(12): 1629-38, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21051460

RESUMEN

More than 25 loci have been linked to type 1 diabetes (T1D) in the nonobese diabetic (NOD) mouse, but identification of the underlying genes remains challenging. We describe here the positional cloning of a T1D susceptibility locus, Idd11, located on mouse chromosome 4. Sequence analysis of a series of congenic NOD mouse strains over a critical 6.9-kb interval in these mice and in 25 inbred strains identified several haplotypes, including a unique NOD haplotype, associated with varying levels of T1D susceptibility. Haplotype diversity within this interval between congenic NOD mouse strains was due to a recombination hotspot that generated four crossover breakpoints, including one with a complex conversion tract. The Idd11 haplotype and recombination hotspot are located within a predicted gene of unknown function, which exhibits decreased expression in relevant tissues of NOD mice. Notably, it was the recombination hotspot that aided our mapping of Idd11 and confirms that recombination hotspots can create genetic variation affecting a common polygenic disease. This finding has implications for human genetic association studies, which may be affected by the approximately 33,000 estimated hotspots in the genome.


Asunto(s)
Intercambio Genético/genética , Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Biología Computacional , Haplotipos/genética , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
12.
Proc Natl Acad Sci U S A ; 102(43): 15551-6, 2005 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-16223880

RESUMEN

Chronic microbial infections are associated with fibrotic and inflammatory reactions known as granulomas showing similarities to wound-healing and tissue repair processes. We have previously mapped three leishmaniasis susceptibility loci, designated lmr1, -2, and -3, which exert their effect independently of T cell immune responses. Here, we show that the wound repair response is critically important for the rapid cure in murine cutaneous leishmaniasis caused by Leishmania major. Mice congenic for leishmaniasis resistance loci, which cured their lesions more rapidly than their susceptible parents, also expressed differentially genes involved in tissue repair, laid down more ordered collagen fibers, and healed punch biopsy wounds more rapidly. Fibroblast monolayers from these mice repaired in vitro wounds faster, and this process was accelerated by supernatants from infected macrophages. Because these effects are independent of T cell-mediated immunity, we conclude that the rate of wound healing is likely to be an important component of innate immunity involved in resistance to cutaneous leishmaniasis.


Asunto(s)
Leishmaniasis Cutánea/genética , Cicatrización de Heridas/genética , Animales , Femenino , Fibroblastos/fisiología , Predisposición Genética a la Enfermedad , Inmunidad Innata , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/fisiopatología , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Linfocitos T/inmunología
13.
Mamm Genome ; 16(1): 32-40, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15674731

RESUMEN

We describe two new mutations, 153Gso and 154Gso, associated with reciprocal translocations with a common breakpoint in mouse chromosome 6B3 (Mmu6B3). The translocations arose independently in offspring of male mice treated with chlorambucil and glycidamide, respectively. Homozygotes of both mutant stocks display a characteristic gait ataxia with 'foot-patting' behavior; despite their ataxia the mutant animals are healthy, long-lived, and breed normally. Breeding experiments confirmed that 153Gso and 154Gso mutations are allelic, and both fail to complement a known mutation hotfoot (ho), a Mmu6 mutation involving the glutamate receptor gene, Grid2, that is associated with a virtually identical phenotype. Our studies demonstrate that the 153Gso and 154Gso mutations disrupt the Grid2 gene at sites located more than 100 kb apart in intron 6 and intron 4 of the gene, respectively. The occurrence of two independent translocations from a relatively small colony within the same locus supports data suggesting the hypermutability of the Grid2 locus and suggest that the gene's large size make it an especially likely target for mutations involving genetic rearrangement.


Asunto(s)
Receptores de Glutamato/genética , Translocación Genética , Animales , Secuencia de Bases , Masculino , Ratones , Ratones Mutantes , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Hum Mol Genet ; 13(22): 2813-21, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15385447

RESUMEN

Increased susceptibility to gastric cancer has been associated with a wide range of host genetic and environmental factors, including Helicobacter pylori infection. Helicobacter pylori infection is postulated to initiate a progression through atrophic gastritis, metaplasia and dysplasia to cancer, and has been associated with reduction of acid output and dysregulation of stomach mucins. Here, we present the characterization of two mouse lines carrying mutant alleles of the gene encoding the Kcnq1 potassium channel, which very rapidly establish chronic gastritis in a pathogen-exposed environment. These mice develop gastric hyperplasia, hypochlorhydria and mucin dysregulation independent of infection. Metaplasia, dysplasia and pre-malignant adenomatous hyperplasia of the stomach have been observed in these Kcnq1 mutant mice, also independent of infection. The data presented here suggest that Kcnq1 mutant mice can be used both as an efficient model for the development of atrophic gastritis after infection and to determine the processes during the later stages of progression to gastric cancer independent of infection. Thus, Kcnq1 mutant mice are a powerful new tool for investigating the connection between acid balance, Helicobacter infection and mucin disruption in the progression to gastric cancer.


Asunto(s)
Adenoma/genética , Gastritis/genética , Canales de Potasio KCNQ/genética , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Aclorhidria/etiología , Aclorhidria/genética , Aclorhidria/patología , Adenoma/patología , Alelos , Animales , Enfermedad Crónica , Femenino , Mucinas Gástricas/metabolismo , Gastritis/etiología , Gastritis/patología , Predisposición Genética a la Enfermedad , Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Hiperplasia , Masculino , Metaplasia , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Miocardio/patología , Lesiones Precancerosas/patología , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Estría Vascular/patología , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...