Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 151(5): 951-63, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23178118

RESUMEN

The inactive X chromosome's (Xi) physical territory is microscopically devoid of transcriptional hallmarks and enriched in silencing-associated modifications. How these microscopic signatures relate to specific Xi sequences is unknown. Therefore, we profiled Xi gene expression and chromatin states at high resolution via allele-specific sequencing in mouse trophoblast stem cells. Most notably, X-inactivated transcription start sites harbored distinct epigenetic signatures relative to surrounding Xi DNA. These sites displayed H3-lysine27-trimethylation enrichment and DNaseI hypersensitivity, similar to autosomal Polycomb targets, yet excluded Pol II and other transcriptional hallmarks, similar to nontranscribed genes. CTCF bound X-inactivated and escaping genes, irrespective of measured chromatin boundaries. Escape from X inactivation occurred within, and X inactivation was maintained exterior to, the area encompassed by Xist in cells subject to imprinted and random X inactivation. The data support a model whereby inactivation of specific regulatory elements, rather than a simple chromosome-wide separation from transcription machinery, governs gene silencing over the Xi.


Asunto(s)
Silenciador del Gen , Elementos Reguladores de la Transcripción , Inactivación del Cromosoma X , Animales , Factor de Unión a CCCTC , Cromatina/metabolismo , Desoxirribonucleasa I/metabolismo , Código de Histonas , Elementos de Nucleótido Esparcido Largo , Ratones , Proteínas del Grupo Polycomb/metabolismo , ARN Polimerasa II/metabolismo , Proteínas Represoras/metabolismo , Células Madre/citología , Células Madre/metabolismo , Trofoblastos/citología
2.
J Am Soc Nephrol ; 28(6): 1741-1752, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28062569

RESUMEN

Ischemia-reperfusion injury (IRI) is a leading cause of AKI. This common clinical complication lacks effective therapies and can lead to the development of CKD. The αvß5 integrin may have an important role in acute injury, including septic shock and acute lung injury. To examine its function in AKI, we utilized a specific function-blocking antibody to inhibit αvß5 in a rat model of renal IRI. Pretreatment with this anti-αvß5 antibody significantly reduced serum creatinine levels, diminished renal damage detected by histopathologic evaluation, and decreased levels of injury biomarkers. Notably, therapeutic treatment with the αvß5 antibody 8 hours after IRI also provided protection from injury. Global gene expression profiling of post-ischemic kidneys showed that αvß5 inhibition affected established injury markers and induced pathway alterations previously shown to be protective. Intravital imaging of post-ischemic kidneys revealed reduced vascular leak with αvß5 antibody treatment. Immunostaining for αvß5 in the kidney detected evident expression in perivascular cells, with negligible expression in the endothelium. Studies in a three-dimensional microfluidics system identified a pericyte-dependent role for αvß5 in modulating vascular leak. Additional studies showed αvß5 functions in the adhesion and migration of kidney pericytes in vitro Initial studies monitoring renal blood flow after IRI did not find significant effects with αvß5 inhibition; however, future studies should explore the contribution of vasomotor effects. These studies identify a role for αvß5 in modulating injury-induced renal vascular leak, possibly through effects on pericyte adhesion and migration, and reveal αvß5 inhibition as a promising therapeutic strategy for AKI.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Riñón/irrigación sanguínea , Receptores de Vitronectina/antagonistas & inhibidores , Daño por Reperfusión/prevención & control , Animales , Masculino , Ratas , Ratas Sprague-Dawley
3.
Nucleic Acids Res ; 42(8): e68, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24561615

RESUMEN

The ability to correlate chromosome conformation and gene expression gives a great deal of information regarding the strategies used by a cell to properly regulate gene activity. 4C-Seq is a relatively new and increasingly popular technology where the set of genomic interactions generated by a single point in the genome can be determined. 4C-Seq experiments generate large, complicated data sets and it is imperative that signal is properly distinguished from noise. Currently, there are a limited number of methods for analyzing 4C-Seq data. Here, we present a new method, fourSig, which in addition to being precise and simple to use also includes a new feature that prioritizes detected interactions. Our results demonstrate the efficacy of fourSig with previously published and novel 4C-Seq data sets and show that our significance prioritization correlates with the ability to reproducibly detect interactions among replicates.


Asunto(s)
Cromosomas/química , Programas Informáticos , Alelos , Animales , Interpretación Estadística de Datos , Expresión Génica , Sitios Genéticos , Genómica/métodos , Hibridación Fluorescente in Situ , Ratones , Conformación de Ácido Nucleico , Globinas beta/genética
4.
Development ; 139(12): 2130-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22573614

RESUMEN

Proper regulation of X-linked gene expression, termed dosage compensation, is required for the normal development of mammalian embryos. Through the process of X chromosome inactivation (XCI), somatic cells of mammalian females inactivate one of their two X chromosomes in order to balance X-linked gene dosage with their male counterparts. The process of XCI is dependent upon the long non-coding RNA Xist, which is expressed from and coats the inactivated X chromosome (Xi) in cis. During mouse embryogenesis, imprinted XCI inactivates the paternally inherited X chromosome (Xp) within the extra-embryonic lineages. Consequently, females harboring a paternally derived Xist mutation (X/X(Xist-)) die owing to failure of imprinted XCI and, presumably, poor trophoblast development. Here, we investigate the consequence of two active X chromosomes in the extra-embryonic ectoderm (ExE) of X/X(Xist-) female embryos. At embryonic day (E) 6.5, we find that the X/X(Xist-) ExE lacks the transcriptional regulator CDX2, a factor required to maintain the ExE in a progenitor state. In addition, spongiotrophoblast progenitors are not maintained. Surprisingly, we observe evidence of an Xi in a subpopulation of X/X(Xist-) ExE cells. We demonstrate further that trophectodermal stem cells derived from X/X(Xist-) embryos completely reverse normal imprinted XCI patterns. Taken together, our data suggest that, much like in the cells of the epiblast, the initial imprint that establishes imprinted XCI is probably erased in ExE cells. Conversely, unlike the epiblast, in which XCI is not required for progenitor cell maintenance, we demonstrate that dosage compensation is indispensable for the maintenance of trophoblast progenitors.


Asunto(s)
Compensación de Dosificación (Genética) , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Membranas Extraembrionarias/citología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Factor de Transcripción CDX2 , Recuento de Células , Ectodermo/citología , Ectodermo/metabolismo , Femenino , Impresión Genómica/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , ARN Largo no Codificante , ARN no Traducido , Factores de Transcripción/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Cromosoma X/genética , Inactivación del Cromosoma X/genética
5.
Dev Biol ; 333(2): 312-23, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19591821

RESUMEN

The functional unit of the kidney is the nephron. During its organogenesis, the mammalian metanephric kidney generates thousands of nephrons over a protracted period of fetal life. All nephrons are derived from a population of self-renewing multi-potent progenitor cells, termed the cap mesenchyme. However, our understanding of the molecular and cellular mechanisms underlying nephron development is at an early stage. In order to identify factors involved in nephrogenesis, we performed a high-resolution, spatial profiling of a number of transcriptional regulators expressed within the cap mesenchyme and early developing nephron. Our results demonstrate novel, stereotypic, spatially defined cellular sub-domains within the cap mesenchyme, which may, in part, reflect induction of nephron precursors. These results suggest a hitherto unappreciated complexity of cell states that accompany the assembly of the metanephric kidney, likely reflecting diverse regulatory actions such as the maintenance and induction of nephron progenitors.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Nefronas/embriología , Animales , Perfilación de la Expresión Génica , Hibridación in Situ , Riñón/embriología , Ratones , Microscopía Fluorescente/métodos , Modelos Biológicos , Nefronas/fisiología , Estructura Terciaria de Proteína , Transducción de Señal , Células Madre/citología , Factores de Tiempo , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo
6.
Dev Biol ; 319(2): 396-405, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18485340

RESUMEN

The mammalian kidney consists of an array of tubules connected to a ductal system that collectively function to control water/salt balance and to remove waste from the organisms' circulatory system. During mammalian embryogenesis, three kidney structures form within the intermediate mesoderm. The two most anterior structures, the pronephros and the mesonephros, are transitory and largely non-functional, while the most posterior, the metanephros, persists as the adult kidney. We have explored the mechanisms underlying regional specific differentiation of the kidney forming mesoderm. Previous studies have shown a requirement for Hox11 paralogs (Hoxa11, Hoxc11 and Hoxd11) in metanephric development. Mice lacking all Hox11 activity fail to form metanephric kidney structures. We demonstrate that the Hox11 paralog expression is restricted in the intermediate mesoderm to the posterior, metanephric level. When Hoxd11 is ectopically activated in the anterior mesonephros, we observe a partial transformation to a metanephric program of development. Anterior Hoxd11(+) cells activate Six2, a transcription factor required for the maintenance of metanephric tubule progenitors. Additionally, Hoxd11(+) mesonephric tubules exhibit an altered morphology and activate several metanephric specific markers normally confined to distal portions of the functional nephron. Collectively, our data support a model where Hox11 paralogs specify a metanephric developmental program in responsive intermediate mesoderm. This program maintains tubule forming progenitors and instructs a metanephric specific pattern of nephron differentiation.


Asunto(s)
Proteínas de Homeodominio/genética , Riñón/embriología , Mesodermo/fisiología , Factores de Transcripción/genética , Animales , Embrión de Mamíferos , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Genotipo , Hibridación in Situ , Túbulos Renales/embriología , Mesonefro/embriología , Ratones
7.
Dev Biol ; 324(1): 88-98, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18835385

RESUMEN

The mammalian metanephric kidney is derived from the intermediate mesoderm. In this report, we use molecular fate mapping to demonstrate that the majority of cell types within the metanephric kidney arise from an Osr1(+) population of metanephric progenitor cells. These include the ureteric epithelium of the collecting duct network, the cap mesenchyme and its nephron epithelia derivatives, the interstitial mesenchyme, vasculature and smooth muscle. Temporal fate mapping shows a progressive restriction of Osr1(+) cell fates such that at the onset of active nephrogenesis, Osr1 activity is restricted to the Six2(+) cap mesenchyme nephron progenitors. However, low-level labeling of Osr1(+) cells suggests that the specification of interstitial mesenchyme and cap mesenchyme progenitors occurs within the Osr1(+) population prior to the onset of metanephric development. Furthermore, although Osr1(+) progenitors give rise to much of the kidney, Osr1 function is only essential for the development of the nephron progenitor compartment. These studies provide new insights into the cellular origins of metanephric kidney structures and lend support to a model where Osr1 function is limited to establishing the nephron progenitor pool.


Asunto(s)
Riñón/embriología , Mesodermo/embriología , Nefronas/embriología , Células Madre/citología , Factores de Transcripción/biosíntesis , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Proteínas de Homeodominio/metabolismo , Riñón/citología , Riñón/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Mutantes , Nefronas/citología , Nefronas/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Urotelio/embriología , Urotelio/metabolismo
8.
Protein Eng Des Sel ; 30(5): 359-372, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28180900

RESUMEN

Wnt signaling pathways are required for a wide variety of biological processes ranging from embryonic development to tissue repair and regeneration. Dickkopf-2 (DKK2) is classically defined as a canonical Wnt inhibitor, though it may play a role in activating non-canonical Wnt pathways in the context of endothelial network formation after acute injury. Here we report the discovery of a fusion partner for a DKK2 polypeptide that significantly improves the expression, biochemical properties and pharmacokinetics (PK) of the DKK2 polypeptide. Specifically, human serum albumin (HSA) was identified as a highly effective fusion partner. Substitution of selected amino acid residues in DKK2 designed to decrease heparan sulfate binding by HSA-DKK2 variants, further improved the PK properties of the molecule in rodents. The HSA-DKK2 variants were monomeric, as thermally stable as wild type, and active as measured by their ability to bind to and prevent phosphorylation of the Wnt coreceptor LRP6. Our engineering efforts resulted in potent long-lived variants of the canonical Wnt inhibitor DKK2, applicable for Wnt pathway manipulation either by systematic delivery or focused administration at sites of tissue injury.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/antagonistas & inhibidores , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión , Albúmina Sérica , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , Animales , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/aislamiento & purificación , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica/biosíntesis , Albúmina Sérica/química , Albúmina Sérica/aislamiento & purificación , Albúmina Sérica/farmacología
9.
Stem Cell Reports ; 3(4): 650-62, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25358792

RESUMEN

The mammalian kidney is a complex organ consisting of multiple cell types. We previously showed that the Six2-expressing cap mesenchyme is a multipotent self-renewing progenitor population for the main body of the nephron, the basic functional unit of the kidney. However, the cellular mechanisms establishing stromal tissues are less clear. We demonstrate that the Foxd1-expressing cortical stroma represents a distinct multipotent self-renewing progenitor population that gives rise to stromal tissues of the interstitium, mesangium, and pericytes throughout kidney organogenesis. Fate map analysis of Foxd1-expressing cells demonstrates that a small subset of these cells contributes to Six2-expressing cells at the early stage of kidney outgrowth. Thereafter, there appears to be a strict nephron and stromal lineage boundary derived from Six2-expressing and Foxd1-expressing cell types, respectively. Taken together, our observations suggest that distinct multipotent self-renewing progenitor populations coordinate cellular differentiation of the nephron epithelium and renal stroma during mammalian kidney organogenesis.


Asunto(s)
Linaje de la Célula , Células Madre Embrionarias/citología , Riñón/citología , Células del Estroma/citología , Animales , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Riñón/embriología , Ratones , Ratones Endogámicos C57BL , Células del Estroma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Genetics ; 197(2): 715-23, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24653000

RESUMEN

X chromosome inactivation (XCI) is an epigenetic process that almost completely inactivates one of two X chromosomes in somatic cells of mammalian females. A few genes are known to escape XCI and the mechanism for this escape remains unclear. Here, using mouse trophoblast stem (TS) cells, we address whether particular chromosomal interactions facilitate escape from imprinted XCI. We demonstrate that promoters of genes escaping XCI do not congregate to any particular region of the genome in TS cells. Further, the escape status of a gene was uncorrelated with the types of genomic features and gene activity located in contacted regions. Our results suggest that genes escaping imprinted XCI do so by using the same regulatory sequences as their expressed alleles on the active X chromosome. We suggest a model where regulatory control of escape from imprinted XCI is mediated by genomic elements located in close linear proximity to escaping genes.


Asunto(s)
Células Madre Embrionarias/citología , Trofoblastos/citología , Inactivación del Cromosoma X , Alelos , Animales , Elementos de Facilitación Genéticos , Femenino , Impresión Genómica , Ratones , Cromosoma X
12.
Cell Stem Cell ; 3(2): 169-81, 2008 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-18682239

RESUMEN

Nephrons, the basic functional units of the kidney, are generated repetitively during kidney organogenesis from a mesenchymal progenitor population. Which cells within this pool give rise to nephrons and how multiple nephron lineages form during this protracted developmental process are unclear. We demonstrate that the Six2-expressing cap mesenchyme represents a multipotent nephron progenitor population. Six2-expressing cells give rise to all cell types of the main body of the nephron during all stages of nephrogenesis. Pulse labeling of Six2-expressing nephron progenitors at the onset of kidney development suggests that the Six2-expressing population is maintained by self-renewal. Clonal analysis indicates that at least some Six2-expressing cells are multipotent, contributing to multiple domains of the nephron. Furthermore, Six2 functions cell autonomously to maintain a progenitor cell status, as cap mesenchyme cells lacking Six2 activity contribute to ectopic nephron tubules, a mechanism dependent on a Wnt9b inductive signal. Taken together, our observations suggest that Six2 activity cell-autonomously regulates a multipotent nephron progenitor population.


Asunto(s)
Proteínas de Homeodominio/genética , Riñón/citología , Nefronas/citología , Organogénesis/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Quimera/genética , Quimera/metabolismo , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Riñón/embriología , Riñón/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Nefronas/embriología , Nefronas/fisiología , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas Wnt/genética
13.
Cell Stem Cell ; 2(3): 284-91, 2008 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-18371453

RESUMEN

Understanding the mechanisms of nephron repair is critical for the design of new therapeutic approaches to treat kidney disease. The kidney can repair after even a severe insult, but whether adult stem or progenitor cells contribute to epithelial renewal after injury and the cellular origin of regenerating cells remain controversial. Using genetic fate-mapping techniques, we generated transgenic mice in which 94%-95% of tubular epithelial cells, but no interstitial cells, were labeled with either beta-galactosidase (lacZ) or red fluorescent protein (RFP). Two days after ischemia-reperfusion injury (IRI), 50.5% of outer medullary epithelial cells coexpress Ki67 and RFP, indicating that differentiated epithelial cells that survived injury undergo proliferative expansion. After repair was complete, 66.9% of epithelial cells had incorporated BrdU, compared to only 3.5% of cells in the uninjured kidney. Despite this extensive cell proliferation, no dilution of either cell-fate marker was observed after repair. These results indicate that regeneration by surviving tubular epithelial cells is the predominant mechanism of repair after ischemic tubular injury in the adult mammalian kidney.


Asunto(s)
Células Madre Adultas/metabolismo , Proliferación Celular , Células Epiteliales/metabolismo , Enfermedades Renales/metabolismo , Túbulos Renales/metabolismo , Regeneración , Daño por Reperfusión/metabolismo , Células Madre Adultas/citología , Animales , Células Epiteliales/patología , Enfermedades Renales/patología , Túbulos Renales/patología , Ratones , Ratones Transgénicos , Daño por Reperfusión/patología , Factores de Tiempo
14.
Dev Biol ; 303(2): 772-83, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17125762

RESUMEN

Members of the ETS family of transcription factors are among the first genes expressed in the developing vasculature, but loss-of-function experiments for individual ETS factors in mice have not uncovered important early functional roles for these genes. However, multiple ETS factors are expressed in spatially and temporally overlapping patterns in the developing vasculature, suggesting possible functional overlap. We have taken a comprehensive approach to exploring the function of these factors during vascular development by employing the genetic and experimental tools available in the zebrafish to analyze four ETS family members expressed together in the zebrafish vasculature; fli1, fli1b, ets1, and etsrp. We isolated and characterized an ENU-induced mutant with defects in trunk angiogenesis and positionally cloned the defective gene from this mutant, etsrp. Using the etsrp morpholinos targeting each of the four genes, we show that the four ETS factors function combinatorially during vascular and hematopoietic development. Reduction of etsrp or any of the other genes alone results in either partial or no defects in endothelial differentiation, while combined reduction in the function of all four genes causes dramatic loss of endothelial cells. Our results demonstrate that combinatorial ETS factor function is essential for early endothelial specification and differentiation.


Asunto(s)
Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Mapeo Cromosómico , Clonación Molecular , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Morfogénesis/genética , Mutación , Neovascularización Fisiológica/genética , Oligodesoxirribonucleótidos Antisentido/genética , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
15.
Genes Dev ; 17(11): 1346-51, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12782653

RESUMEN

In this study, we utilize transgenic zebrafish with fluorescently labeled blood vessels to identify and characterize a mutant (y10) that displays specific defects in the formation of arteries, but not veins. We find that y10 encodes phospholipase C gamma-1 (plcg1), a known effector of receptor tyrosine kinase signaling. We further show that plcg1y10 mutant embryos fail to respond to exogenous Vegf. Our results indicate that Plcg1 functions specifically downstream of the Vegf receptor during embryonic development to govern formation of the arterial system.


Asunto(s)
Arterias/embriología , Factores de Crecimiento Endotelial/genética , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Linfocinas/genética , Fosfolipasas de Tipo C/genética , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Arterias/citología , Secuencia de Bases , Cruzamientos Genéticos , Cartilla de ADN , Embrión no Mamífero/fisiología , Femenino , Tamización de Portadores Genéticos , Masculino , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Músculo Liso Vascular/embriología , Fosfolipasa C gamma , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Proteínas Recombinantes/metabolismo , Fosfolipasas de Tipo C/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA