Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell ; 174(2): 285-299.e12, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29887374

RESUMEN

Visceral adipose tissue (VAT) hosts a population of regulatory T (Treg) cells, with a unique phenotype, that controls local and systemic inflammation and metabolism. Generation of a T cell receptor transgenic mouse line, wherein VAT Tregs are highly enriched, facilitated study of their provenance, dependencies, and activities. We definitively established a role for T cell receptor specificity, uncovered an unexpected function for the primordial Treg transcription-factor, Foxp3, evidenced a cell-intrinsic role for interleukin-33 receptor, and ordered these dependencies within a coherent scenario. Genesis of the VAT-Treg phenotype entailed a priming step in the spleen, permitting them to exit the lymphoid organs and surveil nonlymphoid tissues, and a final diversification process within VAT, in response to microenvironmental cues. Understanding the principles of tissue-Treg biology is a prerequisite for precision-targeting strategies.


Asunto(s)
Grasa Intraabdominal/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T Reguladores/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Factores de Transcripción Forkhead/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Grasa Intraabdominal/inmunología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , PPAR gamma/genética , PPAR gamma/metabolismo , Fenotipo , ARN/química , ARN/aislamiento & purificación , ARN/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina/metabolismo , Análisis de la Célula Individual , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Transcriptoma
2.
Cell ; 162(1): 33-44, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26140591

RESUMEN

SNPs affecting disease risk often reside in non-coding genomic regions. Here, we show that SNPs are highly enriched at mouse strain-selective adipose tissue binding sites for PPARγ, a nuclear receptor for anti-diabetic drugs. Many such SNPs alter binding motifs for PPARγ or cooperating factors and functionally regulate nearby genes whose expression is strain selective and imbalanced in heterozygous F1 mice. Moreover, genetically determined binding of PPARγ accounts for mouse strain-specific transcriptional effects of TZD drugs, providing proof of concept for personalized medicine related to nuclear receptor genomic occupancy. In human fat, motif-altering SNPs cause differential PPARγ binding, provide a molecular mechanism for some expression quantitative trait loci, and are risk factors for dysmetabolic traits in genome-wide association studies. One PPARγ motif-altering SNP is associated with HDL levels and other metabolic syndrome parameters. Thus, natural genetic variation in PPARγ genomic occupancy determines individual disease risk and drug response.


Asunto(s)
Hipoglucemiantes/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Polimorfismo de Nucleótido Simple , Tejido Adiposo , Animales , Expresión Génica , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Factores de Transcripción/metabolismo
3.
Genes Dev ; 32(15-16): 1035-1044, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30006480

RESUMEN

The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is known to regulate lipid metabolism in many tissues, including macrophages. Here we report that peritoneal macrophage respiration is enhanced by rosiglitazone, an activating PPARγ ligand, in a PPARγ-dependent manner. Moreover, PPARγ is required for macrophage respiration even in the absence of exogenous ligand. Unexpectedly, the absence of PPARγ dramatically affects the oxidation of glutamine. Both glutamine and PPARγ have been implicated in alternative activation (AA) of macrophages, and PPARγ was required for interleukin 4 (IL4)-dependent gene expression and stimulation of macrophage respiration. Indeed, unstimulated macrophages lacking PPARγ contained elevated levels of the inflammation-associated metabolite itaconate and express a proinflammatory transcriptome that, remarkably, phenocopied that of macrophages depleted of glutamine. Thus, PPARγ functions as a checkpoint, guarding against inflammation, and is permissive for AA by facilitating glutamine metabolism. However, PPARγ expression is itself markedly increased by IL4. This suggests that PPARγ functions at the center of a feed-forward loop that is central to AA of macrophages.


Asunto(s)
Glutamina/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , PPAR gamma/fisiología , Animales , Respiración de la Célula , Células Cultivadas , Ácidos Grasos/metabolismo , Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Interleucina-4/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/genética , Rosiglitazona , Tiazolidinedionas/farmacología
4.
Semin Immunol ; 27(5): 315-21, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26616665

RESUMEN

Adipose tissue is composed of many functionally and developmentally distinct cell types, the metabolic core of which is the adipocyte. The classification of "adipocyte" encompasses three primary types - white, brown, and beige - with distinct origins, anatomic distributions, and homeostatic functions. The ability of adipocytes to store and release lipids, respond to insulin, and perform their endocrine functions (via secretion of adipokines) is heavily influenced by the immune system. Various cell populations of the innate and adaptive arms of the immune system can resist or exacerbate the development of the chronic, low-grade inflammation associated with obesity and metabolic dysfunction. Here, we discuss these interactions, with a focus on their consequences for adipocyte and adipose tissue function in the setting of chronic overnutrition. In addition, we will review the effects of diet composition on adipose tissue inflammation and recent evidence suggesting that diet-driven disruption of the gut microbiota can trigger pathologic inflammation of adipose tissue.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Homeostasis , Inmunidad , Adipocitos/metabolismo , Tejido Adiposo/patología , Animales , Comunicación Celular , Dieta , Elasticidad , Metabolismo Energético , Microbioma Gastrointestinal/inmunología , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Obesidad/complicaciones , Obesidad/inmunología , Obesidad/metabolismo , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 110(46): 18656-61, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24167256

RESUMEN

Adipose tissue is an important metabolic organ, the dysfunction of which is associated with the development of obesity, diabetes mellitus, and cardiovascular disease. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is considered the master regulator of adipocyte differentiation and function. Although its cell-autonomous role in adipogenesis has been clearly demonstrated in cell culture, previous fat-specific knockouts of the murine PPARγ gene did not demonstrate a dramatic phenotype in vivo. Here, using Adipoq-Cre mice to drive adipose-specific recombination, we report a unique fat-specific PPARγ knockout (PPARγ FKO) mouse model with almost no visible brown and white adipose tissue at age 3 mo. As a consequence, PPARγ FKO mice had hugely enlarged pancreatic islets, massive fatty livers, and dramatically elevated levels of blood glucose and serum insulin accompanied by extreme insulin resistance. PPARγ FKO mice also exhibited delayed hair coat formation associated with absence of dermal fat, disrupted mammary gland development with loss of mammary fat pads, and high bone mass with loss of bone marrow fat, indicating the critical roles of adipose PPARγ in these tissues. Together, our data reveal the necessity of fat PPARγ in adipose formation, whole-body metabolic homeostasis, and normal development of fat-containing tissues.


Asunto(s)
Adipocitos/metabolismo , Resistencia a la Insulina/genética , Obesidad/metabolismo , PPAR gamma/deficiencia , Animales , Compuestos Azo , Immunoblotting , Inmunohistoquímica , Resistencia a la Insulina/fisiología , Ratones , PPAR gamma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microtomografía por Rayos X
6.
Mol Ther ; 22(9): 1698-706, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24861055

RESUMEN

Functional exhaustion of antigen-specific T cells is a defining characteristic of many chronic infections, but the underlying mechanisms of T cell dysfunction are not well understood. Epigenetics plays an important role in the control of T cell development, differentiation, and function. To examine if epigenetics also plays a role in T cell exhaustion, we analyzed chromatin remodeling in CD8(+) T cells from mice with chronic lymphocytic choriomeningitis virus infection. We observed downregulation of diacetylated histone H3 in both virus-specific and total CD8(+) T cells, and functional defects not only in virus-specific CD8(+) T cells but also within the total CD8(+) T cell population. In vitro treatment of these exhausted CD8(+) T cells with histone deacetylase inhibitors restored diacetylated histone H3 levels, and improved their immune functions. Upon adoptive transfer, these treated CD8(+) T cells developed into functional memory T cells in vivo that enhanced protective immunity. These results define a role of epigenetics in T cell exhaustion and suggest epigenetic manipulation as a novel molecular therapy to restore immune functions.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/metabolismo , Epigénesis Genética , Coriomeningitis Linfocítica/veterinaria , Virus de la Coriomeningitis Linfocítica/fisiología , Traslado Adoptivo , Animales , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/virología , Ensamble y Desensamble de Cromatina , Inhibidores Enzimáticos/farmacología , Femenino , Histonas/metabolismo , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL
7.
J Immunol ; 184(9): 4631-6, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20308634

RESUMEN

Following stimulation, memory T (T(M)) cells rapidly express many effector functions, a hallmark feature that allows them to provide protective immunity. Recent studies suggest that genes involved in this rapid recall response may maintain an open chromatin structure in resting T(M) cells via epigenetic modifications. However, these studies have mostly focused on a few loci, and the techniques used required a large number of cells. We have developed a flow cytometric assay measuring histone modifications in individual murine T cells in combination with lineage-specific markers. In this study, we show that the per-cell level of a marker of open chromatin, diacetylated histone H3 (diAcH3), increases as naive CD8(+) T cells develop into T(M) cells, demonstrating a novel correlation between the differentiation state of a CD8(+) T cell and its abundance of a specific histone modification. Furthermore, our results show that T(M) cells defective in rapid recall ability have less diAcH3 than their fully functional counterparts, indicating that the diAcH3 level of individual T(M) cells is a useful marker for assessing their functionality.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Histonas/metabolismo , Memoria Inmunológica , Subgrupos de Linfocitos T/metabolismo , Acetilación , Animales , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/virología , Diferenciación Celular/genética , Células Cultivadas , Femenino , Citometría de Flujo/métodos , Memoria Inmunológica/genética , Inmunofenotipificación , Listeria monocytogenes/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Procesamiento Proteico-Postraduccional/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/microbiología , Subgrupos de Linfocitos T/virología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/microbiología , Linfocitos T Citotóxicos/virología
8.
Cell Metab ; 32(4): 665-675.e6, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32941798

RESUMEN

The thiazolidinediones (TZDs) are ligands of PPARγ that improve insulin sensitivity, but their use is limited by significant side effects. Recently, we demonstrated a mechanism wherein TZDs improve insulin sensitivity distinct from receptor agonism and adipogenesis: reversal of obesity-linked phosphorylation of PPARγ at serine 273. However, the role of this modification hasn't been tested genetically. Here we demonstrate that mice encoding an allele of PPARγ that cannot be phosphorylated at S273 are protected from insulin resistance, without exhibiting differences in body weight or TZD-associated side effects. Indeed, hyperinsulinemic-euglycemic clamp experiments confirm insulin sensitivity. RNA-seq in these mice reveals reduced expression of Gdf3, a BMP family member. Ectopic expression of Gdf3 is sufficient to induce insulin resistance in lean, healthy mice. We find Gdf3 inhibits BMP signaling and insulin signaling in vitro. Together, these results highlight the diabetogenic role of PPARγ S273 phosphorylation and focus attention on a putative target, Gdf3.


Asunto(s)
Factor 3 de Diferenciación de Crecimiento/metabolismo , Obesidad/tratamiento farmacológico , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Alelos , Animales , Células Cultivadas , Factor 3 de Diferenciación de Crecimiento/genética , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , PPAR gamma/genética , Fosforilación/efectos de los fármacos
9.
Sci Immunol ; 3(27)2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30217811

RESUMEN

Foxp3+CD4+ regulatory T cells (Tregs) accumulate in certain nonlymphoid tissues, where they control diverse aspects of organ homeostasis. Populations of tissue Tregs, as they have been termed, have transcriptomes distinct from those of their counterparts in lymphoid organs and other nonlymphoid tissues. We examined the diversification of Tregs in visceral adipose tissue, skeletal muscle, and the colon vis-à-vis lymphoid organs from the same individuals. The unique transcriptomes of the various tissue Treg populations resulted from layering of tissue-restricted open chromatin regions over regions already open in the spleen, the latter tagged by super-enhancers and particular histone marks. The binding motifs for a small number of transcription factor (TF) families were repeatedly enriched within the accessible chromatin stretches of Tregs in the three nonlymphoid tissues. However, a bioinformatically and experimentally validated transcriptional network, constructed by integrating chromatin accessibility and single-cell transcriptomic data, predicted reliance on different TF family members in the different tissues. The network analysis also revealed that tissue-restricted and broadly acting TFs were integrated into feed-forward loops to enforce tissue-specific gene expression in nonlymphoid-tissue Tregs. Overall, this study provides a framework for understanding the epigenetic dynamics of T cells operating in nonlymphoid tissues, which should inform strategies for specifically targeting them.


Asunto(s)
Colon/inmunología , Grasa Intraabdominal/inmunología , Músculo Esquelético/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/inmunología , Perfilación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Análisis de la Célula Individual
10.
J Clin Invest ; 127(4): 1451-1462, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28240605

RESUMEN

Obesity causes insulin resistance, and PPARγ ligands such as rosiglitazone are insulin sensitizing, yet the mechanisms remain unclear. In C57BL/6 (B6) mice, obesity induced by a high-fat diet (HFD) has major effects on visceral epididymal adipose tissue (eWAT). Here, we report that HFD-induced obesity in B6 mice also altered the activity of gene regulatory elements and genome-wide occupancy of PPARγ. Rosiglitazone treatment restored insulin sensitivity in obese B6 mice, yet, surprisingly, had little effect on gene expression in eWAT. However, in subcutaneous inguinal fat (iWAT), rosiglitazone markedly induced molecular signatures of brown fat, including the key thermogenic gene Ucp1. Obesity-resistant 129S1/SvImJ mice (129 mice) displayed iWAT browning, even in the absence of rosiglitazone. The 129 Ucp1 locus had increased PPARγ binding and gene expression that were preserved in the iWAT of B6x129 F1-intercrossed mice, with an imbalance favoring the 129-derived alleles, demonstrating a cis-acting genetic difference. Thus, B6 mice have genetically defective Ucp1 expression in iWAT. However, when Ucp1 was activated by rosiglitazone, or by iWAT browning in cold-exposed or young mice, expression of the B6 version of Ucp1 was no longer defective relative to the 129 version, indicating epigenomic rescue. These results provide a framework for understanding how environmental influences like drugs can affect the epigenome and potentially rescue genetically determined disease phenotypes.


Asunto(s)
Epigénesis Genética , Obesidad/metabolismo , PPAR gamma/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Hipoglucemiantes/farmacología , Grasa Intraabdominal/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Unión Proteica , Elementos Reguladores de la Transcripción , Rosiglitazona , Grasa Subcutánea Abdominal/metabolismo , Tiazolidinedionas/farmacología , Activación Transcripcional , Transcriptoma , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
11.
Sci Rep ; 6: 27005, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27243788

RESUMEN

Memory T cells (TM) play a prominent role in protection and auto-immunity due to their ability to mount a more effective response than naïve T cells (TN). However, the molecular mechanisms underlying enhanced functionality of TM are not well defined, particularly in human TM. We examined the global gene expression profiles of human CD8(+) TN and TM before and after stimulation. There were 1,284, 1,373 and 1,629 differentially expressed genes between TN and TM at 0 hr, 4 hr and 24 hr after stimulation, respectively, with more genes expressed to higher levels in TM. Genes rapidly up-regulated in TN cells were largely involved in nitrogen, nucleoside and amino acid metabolisms. In contrast, those in CD8(+) TM were significantly enriched for immune-response-associated processes, including cytokine production, lymphocyte activation and chemotaxis. Multiple cytokines were rapidly up-regulated in TM cells, including effector cytokines known to be produced by CD8(+) T cells and important for their functions, as well as regulatory cytokines, both pro- and anti-inflammatory, that are not typically produced by CD8(+) T cells. These results provide new insights into molecular mechanisms that contribute to the enhanced functionality of human CD8(+) TM and their prominent role in protection and auto-immunity.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Regulación de la Expresión Génica/inmunología , Genoma Humano/inmunología , Memoria Inmunológica/genética , Transcriptoma , Aminoácidos/inmunología , Aminoácidos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Quimiotaxis/genética , Quimiotaxis/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Perfilación de la Expresión Génica , Humanos , Activación de Linfocitos , Análisis por Micromatrices , Anotación de Secuencia Molecular , Nitrógeno/inmunología , Nitrógeno/metabolismo , Nucleósidos/inmunología , Nucleósidos/metabolismo , Cultivo Primario de Células
12.
J Mol Endocrinol ; 51(3): T115-40, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24096517

RESUMEN

The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.


Asunto(s)
Receptores Nucleares Huérfanos/metabolismo , Factores de Transcripción/metabolismo , Animales , Humanos
13.
Mol Cell Biol ; 33(16): 3354-64, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23775123

RESUMEN

"Master" transcription factors are the gatekeepers of lineage identity. As such, they have been a major focus of efforts to manipulate cell fate for therapeutic purposes. The ETS transcription factor PU.1 has a potent ability to confer macrophage phenotypes on cells already committed to a different lineage, but how it overcomes the presence of other master regulators is not known. The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is the master regulator of the adipose lineage, and its genomic binding pattern in adipocytes is well characterized. Here we show that, when expressed at macrophage levels in mature adipocytes, PU.1 bound a large fraction of its macrophage sites, where it induced chromatin opening and the expression of macrophage target genes. Strikingly, PU.1 markedly reduced the genomic binding of PPARγ without changing its abundance. PU.1 expression repressed genes with nearby adipocyte-specific PPARγ binding sites, while a common macrophage-adipocyte gene expression program was retained. Together, these data reveal unexpected lability within the adipocyte PPARγ cistrome and show that, even in terminally differentiated cells, PU.1 can remodel the cistrome of another master regulator.


Asunto(s)
Adipocitos/metabolismo , Regulación de la Expresión Génica , Macrófagos/metabolismo , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipogénesis , Tejido Adiposo Blanco/metabolismo , Animales , Expresión Génica , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética
14.
Cell Res ; 20(1): 13-23, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20029390

RESUMEN

The functional roles of memory B and T lymphocytes underlie the phenomenal success of prophylactic vaccinations, which have decreased morbidities and mortalities from infectious diseases globally over the last 50 years. However, it is becoming increasingly appreciated that memory cells are also capable of mediating the pathology associated with autoimmune disorders and transplant rejection, and may pose a significant barrier to future clinical advancement in immunoregulation. Therefore, understanding the unique properties of memory lymphocytes (as compared to their naive precursors) is a major area of investigation. Here, we focus on one of those singular properties of memory T cells (T(M))-rapid recall. As will be discussed in more detail, rapid recall refers to the ability of quiescent T(M) cells to efficiently and robustly express 'effector functions' following stimulation. Studies that have advanced our understanding of T(M) cells' rapid recall using CD4(+) T cells have been expertly reviewed elsewhere, so we will focus primarily on studies of CD8(+) T cells. We will first review the different ways that CD8(+) T(M) cells can be generated, followed by discussing how this influences their functional properties in the settings of immune protection and pathology. Then, rapid recall ability will be discussed, with emphasis placed on what is currently known about the mechanisms that underlie this unique property of T(M) cells.


Asunto(s)
Inmunidad Adaptativa/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Inmunidad Adaptativa/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Quimiocinas/biosíntesis , Quimiocinas/genética , Citocinas/biosíntesis , Citocinas/genética , Humanos , Memoria Inmunológica/genética , Activación de Linfocitos/genética , Tiempo de Reacción/genética , Tiempo de Reacción/inmunología , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA