Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Chem Biol ; 19(4): 451-459, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36482094

RESUMEN

Bacteria use two-component system (TCS) signaling pathways to sense and respond to peptides involved in host defense, quorum sensing and inter-bacterial warfare. However, little is known about the broad peptide-sensing capabilities of TCSs. In this study, we developed an Escherichia coli display method to characterize the effects of human antimicrobial peptides (AMPs) on the pathogenesis-regulating TCS PhoPQ of Salmonella Typhimurium with much higher throughput than previously possible. We found that PhoPQ senses AMPs with diverse sequences, structures and biological functions. We further combined thousands of displayed AMP variants with machine learning to identify peptide sub-domains and biophysical features linked to PhoPQ activation. Most of the newfound AMP activators induce PhoPQ in S. Typhimurium, suggesting possible roles in virulence regulation. Finally, we present evidence that PhoPQ peptide-sensing specificity has evolved across commensal and pathogenic bacteria. Our method enables new insights into the specificities, mechanisms and evolutionary dynamics of TCS-mediated peptide sensing in bacteria.


Asunto(s)
Proteínas Bacterianas , Escherichia coli , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Bacterianas/metabolismo , Bacterias/metabolismo , Salmonella typhimurium/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Regulación Bacteriana de la Expresión Génica
2.
Microbiol Spectr ; 10(3): e0061922, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35579463

RESUMEN

Typhoid fever is caused primarily by the enteric microbe Salmonella enterica serovar Typhi and remains a major global health problem with approximately 14 million new infections and 136,000 fatalities annually. While there are antibiotic options available to treat the disease, the global increase in multidrug-resistant strains necessitates alternative therapeutic options. Host-targeted therapeutics present a promising anti-infective strategy against intracellular bacterial pathogens. A cell-based assay identified a compound that inhibits Salmonella proliferation in infected cells, 2-(3-hydroxypropyl)-1-(3-phenoxyphenyl)-1,2-dihydrochromeno[2,3-c]pyrrole-3,9-dione (KH-1), which is devoid of direct activity against Salmonella. The compound inhibits the growth of both antibiotic-sensitive and -resistant Salmonella strains inside macrophages and reduces lactate dehydrogenase (LDH) release from Salmonella-infected cells. Subsequent screening of KH-1 commercial analogs identified 2-(4-fluorobenzyl)-1-(3-phenoxyphenyl)-1,2-dihydrochromeno[2,3-c] pyrrole-3,9-dione (KH-1-2), which is more effective in controlling Salmonella growth inside macrophages. In vitro KH-1-2 treatment of Salmonella infection resulted in an 8- to 10-fold reduction in bacterial load in infected macrophages. In combination with suboptimal ciprofloxacin treatment, KH-1-2 further reduces Salmonella growth inside macrophages. The toxicity and efficacy of KH-1-2 in controlling Salmonella infection were examined in vivo using a mouse model of typhoid fever. No significant compound-related clinical signs and histological findings of the liver, spleen, or kidney were observed from uninfected mice that were intraperitoneally treated with KH-1-2. KH-1-2 significantly protected mice from a lethal dose of infection by an antibiotic-resistant Salmonella strain. Thus, our study provides support that this is a promising lead compound for the development of a novel host-targeted therapeutic agent to control typhoid fever. IMPORTANCESalmonella spp. cause significant morbidity and mortality worldwide. Typhoidal spp. (e.g., S. Typhi) cause a systemic disease typically treated with antibiotics. However, growing antibiotic resistance is resulting in increased treatment failures. We screened a compound library for those that would reduce Salmonella-induced macrophage toxicity, identifying compound KH-1. KH-1 has no direct effects on the bacteria but limits Salmonella survival in macrophages and protects against lethal infection in a mouse model of typhoid fever. A suboptimal concentration of ciprofloxacin worked in conjunction with the compound to further decrease Salmonella survival in macrophages. An analog (KH-1-2) was identified that possessed increased activity in vitro in macrophages and in vivo against both antibiotic-sensitive and -resistant strains. Thus, we report the identification of a lead compound that may be a useful scaffold as a host-directed antimicrobial against typhoid fever.


Asunto(s)
Antiinfecciosos , Fiebre Tifoidea , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/uso terapéutico , Ciprofloxacina/farmacología , Ciprofloxacina/uso terapéutico , Humanos , Pirroles/farmacología , Pirroles/uso terapéutico , Salmonella , Salmonella typhi , Fiebre Tifoidea/tratamiento farmacológico , Fiebre Tifoidea/microbiología , Fiebre Tifoidea/prevención & control
3.
Artículo en Inglés | MEDLINE | ID: mdl-31263682

RESUMEN

Bacteria alter gene expression in response to changes in their environment through various mechanisms that include signal transduction systems. These signal transduction systems use membrane histidine kinase with sensing domains to mediate phosphotransfer to DNA-binding proteins that alter the level of gene expression. Such regulators are called two-component systems (TCSs). TCSs integrate external signals and information from stress pathways, central metabolism and other global regulators, thus playing an important role as part of the overall regulatory network. This review will focus on the knowledge of TCSs in the Gram-negative bacterium, Francisella tularensis, a biothreat agent with a wide range of potential hosts and a significant ability to cause disease. While TCSs have been well-studied in several bacterial pathogens, they have not been well-studied in non-model organisms, such as F. tularensis and its subspecies, whose canonical TCS content surprisingly ranges from few to none. Additionally, of those TCS genes present, many are orphan components, including KdpDE, QseC, QseB/PmrA, and an unnamed two-component system (FTN_1452/FTN_1453). We discuss recent advances in this field related to the role of TCSs in Francisella physiology and pathogenesis and compare the TCS genes present in human virulent versus. environmental species and subspecies of Francisella.


Asunto(s)
Francisella tularensis/genética , Francisella tularensis/metabolismo , Tularemia/microbiología , Animales , Proteínas Bacterianas/genética , Proteínas de Unión al ADN , Francisella tularensis/patogenicidad , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/genética , Histidina Quinasa/metabolismo , Humanos , Dominios Proteicos , Transducción de Señal , Tularemia/transmisión , Virulencia/genética
4.
Mol Pharm ; 15(11): 5336-5348, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30296381

RESUMEN

Previously we have encapsulated host-directed therapy AR-12 into acetalated dextran (Ace-DEX) microparticles (MPs) to mitigate drug toxicity and passively target phagocytic host cells. Herein, we have improved upon our initial emulsion-based formulation of Ace-DEX MPs encapsulating AR-12 (AR-12/MPs) by improving the drug encapsulation efficiency, evaluating sterilization processes for manufacturing, and understanding cellular and in vivo trafficking of the MPs. By using an alternative solvent system, ethyl acetate, we report an increased encapsulation efficiency of AR-12 while maintaining the pH-responsive degradation kinetics of Ace-DEX MPs. To better manufacture this novel antimicrobial formulation, we sterilized AR-12/MPs by gamma irradiation or ethylene oxide and evaluated their efficacy against intracellular Salmonella enterica serovar Typhi. Sterilized AR-12/MPs resulted in a significant reduction in intracellular bacterial burden compared to Blank/MPs. We also characterized intracellular trafficking of Ace-DEX MPs encapsulating fluorophores, which demonstrated internalization of MPs in endo/lysosomal compartments and time and degradation-rate dependent lysosomal escape into cytosolic compartments. Additionally, in vivo toxicity was mitigated following encapsulation of AR-12, where the maximum tolerated dose of AR-12 was increased compared to soluble treatment via intranasal, intravenous, and intraperitoneal administration routes. Following in vivo trafficking of Ace-DEX MPs via the same routes, intranasal administration demonstrated the highest accumulation in the lungs, liver, and kidneys, which persisted out to 240 h. Overall, we have advanced the formulation of this host-directed therapy and broadened the understanding of Ace-DEX MP delivery.


Asunto(s)
Antibacterianos/administración & dosificación , Portadores de Fármacos/química , Pirazoles/administración & dosificación , Salmonella typhi/efectos de los fármacos , Sulfonamidas/administración & dosificación , Fiebre Tifoidea/tratamiento farmacológico , Acetales/química , Animales , Línea Celular , Células Cultivadas , Dextranos/química , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Emulsiones , Femenino , Células Madre Hematopoyéticas , Humanos , Concentración de Iones de Hidrógeno , Macrófagos , Masculino , Ratones , Ratones Endogámicos BALB C , Cultivo Primario de Células , Fiebre Tifoidea/microbiología
5.
PLoS Pathog ; 14(6): e1007100, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29928066

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)γ is a global transcriptional regulator associated with anti-inflammatory actions. It is highly expressed in alveolar macrophages (AMs), which are unable to clear the intracellular pathogen Mycobacterium tuberculosis (M.tb). Although M.tb infection induces PPARγ in human macrophages, which contributes to M.tb growth, the mechanisms underlying this are largely unknown. We undertook NanoString gene expression analysis to identify novel PPARγ effectors that condition macrophages to be more susceptible to M.tb infection. This revealed several genes that are differentially regulated in response to PPARγ silencing during M.tb infection, including the Bcl-2 family members Bax (pro-apoptotic) and Mcl-1 (pro-survival). Apoptosis is an important defense mechanism that prevents the growth of intracellular microbes, including M.tb, but is limited by virulent M.tb. This suggested that M.tb differentially regulates Mcl-1 and Bax expression through PPARγ to limit apoptosis. In support of this, gene and protein expression analysis revealed that Mcl-1 expression is driven by PPARγ during M.tb infection in human macrophages. Further, 15-lipoxygenase (15-LOX) is critical for PPARγ activity and Mcl-1 expression. We also determined that PPARγ and 15-LOX regulate macrophage apoptosis during M.tb infection, and that pre-clinical therapeutics that inhibit Mcl-1 activity significantly limit M.tb intracellular growth in both human macrophages and an in vitro TB granuloma model. In conclusion, identification of the novel PPARγ effector Mcl-1 has determined PPARγ and 15-LOX are critical regulators of apoptosis during M.tb infection and new potential targets for host-directed therapy for M.tb.


Asunto(s)
Apoptosis , Regulación de la Expresión Génica , Macrófagos Alveolares/patología , Mycobacterium tuberculosis/fisiología , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tuberculosis/patología , Células Cultivadas , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , PPAR gamma/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal , Tuberculosis/metabolismo , Tuberculosis/microbiología
6.
Front Immunol ; 9: 561, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29632532

RESUMEN

Francisella tularensis is a remarkably infectious facultative intracellular bacterium of macrophages that causes tularemia. Early evasion of host immune responses contributes to the success of F. tularensis as a pathogen. F. tularensis entry into human monocytes and macrophages is mediated by the major phagocytic receptor, complement receptor 3 (CR3, CD11b/CD18). We recently determined that despite a significant increase in macrophage uptake following C3 opsonization of the virulent Type A F. tularensis spp. tularensis Schu S4, this phagocytic pathway results in limited pro-inflammatory cytokine production. Notably, MAP kinase/ERK activation is suppressed immediately during C3-opsonized Schu S4-CR3 phagocytosis. A mathematical model of CR3-TLR2 crosstalk predicted early involvement of Ras GTPase-activating protein (RasGAP) in immune suppression by CR3. Here, we link CR3-mediated uptake of opsonized Schu S4 by human monocytes and macrophages with inhibition of early signal 1 inflammasome activation, evidenced by limited caspase-1 cleavage and IL-18 release. This inhibition is due to increased RasGAP activity, leading to a reduction in the Ras-ERK signaling cascade upstream of the early inflammasome activation event. Thus, our data uncover a novel signaling pathway mediated by CR3 following engagement of opsonized virulent F. tularensis to limit inflammasome activation in human phagocytic cells, thereby contributing to evasion of the host innate immune system.


Asunto(s)
Francisella tularensis/inmunología , Inflamasomas/inmunología , Antígeno de Macrófago-1/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Proteínas Activadoras de ras GTPasa/inmunología , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Francisella tularensis/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Inflamasomas/metabolismo , Interleucina-18/inmunología , Interleucina-18/metabolismo , Antígeno de Macrófago-1/metabolismo , Macrófagos/microbiología , Monocitos/inmunología , Monocitos/microbiología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo
7.
Front Microbiol ; 8: 1695, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28955308

RESUMEN

Francisella tularensis (F. tularensis) is the causative agent of tularemia and is classified as a Tier 1 select agent. No licensed vaccine is currently available in the United States and treatment of tularemia is confined to few antibiotics. In this study, we demonstrate that AR-13, a derivative of the cyclooxygenase-2 inhibitor celecoxib, exhibits direct in vitro bactericidal killing activity against Francisella including a type A strain of F. tularensis (SchuS4) and the live vaccine strain (LVS), as well as toward the intracellular proliferation of LVS in macrophages, without causing significant host cell toxicity. Identification of an AR-13-resistant isolate indicates that this compound has an intracellular target(s) and that efflux pumps can mediate AR-13 resistance. In the mouse model of tularemia, AR-13 treatment protected 50% of the mice from lethal LVS infection and prolonged survival time from a lethal dose of F. tularensis SchuS4. Combination of AR-13 with a sub-optimal dose of gentamicin protected 60% of F. tularensis SchuS4-infected mice from death. Taken together, these data support the translational potential of AR-13 as a lead compound for the further development of new anti-Francisella agents.

8.
J Am Heart Assoc ; 5(9)2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27620887

RESUMEN

BACKGROUND: Sepsis patients with cardiac dysfunction have significantly higher mortality. Although several pathways are associated with myocardial damage in sepsis, the precise cause(s) remains unclear and treatment options are limited. This study was designed to develop a new model to investigate the early events of cardiac damage during sepsis progression. METHODS AND RESULTS: Francisella tularensis subspecies novicida (Ft.n) is a Gram-negative intracellular pathogen causing severe sepsis syndrome in mice. BALB/c mice (N=12) were sham treated or infected with Ft.n through the intranasal route. Serial electrocardiograms were recorded at multiple time points until 96 hours. Hearts were then harvested for histology and gene expression studies. Similar to septic patients, we illustrate both cardiac electrical and structural phenotypes in our murine Ft.n infection model, including prominent R' wave formation, prolonged QRS intervals, and significant left ventricular dysfunction. Notably, in infected animals, we detected numerous microlesions in the myocardium, previously observed following nosocomial Streptococcus infection and in sepsis patients. We show that Ft.n-mediated microlesions are attributed to cardiomyocyte apoptosis, increased immune cell infiltration, and expression of inflammatory mediators (tumor necrosis factor, interleukin [IL]-1ß, IL-8, and superoxide dismutase 2). Finally, we identify increased expression of microRNA-155 and rapid degradation of heat shock factor 1 following cardiac Ft.n infection as a primary cause of myocardial inflammation and apoptosis. CONCLUSIONS: We have developed and characterized an Ft.n infection model to understand the pathogenesis of cardiac dysregulation in sepsis. Our findings illustrate novel in vivo phenotypes underlying cardiac dysfunction during Ft.n infection with significant translational impact on our understanding of sepsis pathophysiology.


Asunto(s)
Corazón/fisiopatología , Miocardio/patología , Sepsis/fisiopatología , Tularemia/fisiopatología , Animales , Apoptosis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Electrocardiografía , Factores de Transcripción del Choque Térmico/metabolismo , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Ratones , MicroARNs/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/patología , Sepsis/metabolismo , Sepsis/patología , Superóxido Dismutasa/metabolismo , Tularemia/metabolismo , Tularemia/patología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Infect Immun ; 84(11): 3243-3251, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27600501

RESUMEN

Salmonella enterica serovar Typhi, the causative agent of typhoid fever in humans, forms biofilms encapsulated by an extracellular matrix (ECM). Biofilms facilitate colonization and persistent infection in gallbladders of humans and mouse models of chronic carriage. Individual roles of matrix components have not been completely elucidated in vitro or in vivo To examine individual functions, strains of Salmonella enterica serovar Typhimurium, the murine model of S Typhi, in which various ECM genes were deleted or added, were created to examine biofilm formation, colonization, and persistence in the gallbladder. Studies show that curli contributes most significantly to biofilm formation. Expression of Vi antigen decreased biofilm formation in vitro and virulence and bacterial survival in vivo without altering the examined gallbladder pro- or anti-inflammatory cytokines. Oppositely, loss of all ECM components (ΔwcaM ΔcsgA ΔyihO ΔbcsE) increased virulence and bacterial survival in vivo and reduced gallbladder interleukin-10 (IL-10) levels. Colanic acid and curli mutants had the largest defects in biofilm-forming ability and contributed most significantly to the virulence increase of the ΔwcaM ΔcsgA ΔyihO ΔbcsE mutant strain. While the ΔwcaM ΔcsgA ΔyihO ΔbcsE mutant was not altered in resistance to complement or growth in macrophages, it attached and invaded macrophages better than the wild-type (WT) strain. These data suggest that ECM components have various levels of importance in biofilm formation and gallbladder colonization and that the ECM diminishes disseminated disease in our model, perhaps by reducing cell attachment/invasion and dampening inflammation by maintaining/inducing IL-10 production. Understanding how ECM components aid acute disease and persistence could lead to improvements in therapeutic treatment of typhoid fever patients.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Matriz Extracelular/metabolismo , Vesícula Biliar/microbiología , Salmonella typhimurium/fisiología , Animales , Supervivencia Celular/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Interleucina-10/metabolismo , Macrófagos/microbiología , Ratones , Ratones Transgénicos , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo , Virulencia/fisiología
10.
Antimicrob Agents Chemother ; 60(4): 2052-62, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26787696

RESUMEN

Francisella tularensiscauses tularemia and is a potential biothreat. Given the limited antibiotics for treating tularemia and the possible use of antibiotic-resistant strains as a biowarfare agent, new antibacterial agents are needed. AR-12 is an FDA-approved investigational new drug (IND) compound that induces autophagy and has shown host-directed, broad-spectrum activityin vitroagainstSalmonella entericaserovar Typhimurium andF. tularensis We have shown that AR-12 encapsulated within acetalated dextran (Ace-DEX) microparticles (AR-12/MPs) significantly reduces host cell cytotoxicity compared to that with free AR-12, while retaining the ability to controlS.Typhimurium within infected human macrophages. In the present study, the toxicity and efficacy of AR-12/MPs in controlling virulent type AF. tularensisSchuS4 infection were examinedin vitroandin vivo No significant toxicity of blank MPs or AR-12/MPs was observed in lung histology sections when the formulations were given intranasally to uninfected mice. In histology sections from the lungs of intranasally infected mice treated with the formulations, increased macrophage infiltration was observed for AR-12/MPs, with or without suboptimal gentamicin treatment, but not for blank MPs, soluble AR-12, or suboptimal gentamicin alone. AR-12/MPs dramatically reduced the burden ofF. tularensisin infected human macrophages, in a manner similar to that of free AR-12. However,in vivo, AR-12/MPs significantly enhanced the survival ofF. tularensisSchuS4-infected mice compared to that seen with free AR-12. In combination with suboptimal gentamicin treatment, AR-12/MPs further improved the survival ofF. tularensisSchuS4-infected mice. These studies provide support for Ace-DEX-encapsulated AR-12 as a promising new therapeutic agent for tularemia.


Asunto(s)
Acetales/química , Antibacterianos/farmacología , Dextranos/química , Portadores de Fármacos/síntesis química , Francisella tularensis/efectos de los fármacos , Pirazoles/farmacología , Sulfonamidas/farmacología , Tularemia/tratamiento farmacológico , Administración Intranasal , Animales , Carga Bacteriana/efectos de los fármacos , Composición de Medicamentos , Sinergismo Farmacológico , Femenino , Francisella tularensis/crecimiento & desarrollo , Francisella tularensis/patogenicidad , Gentamicinas/farmacología , Humanos , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Bazo/efectos de los fármacos , Bazo/microbiología , Bazo/patología , Análisis de Supervivencia , Tularemia/microbiología , Tularemia/mortalidad , Tularemia/patología
11.
Int J Pharm ; 477(1-2): 334-43, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25447826

RESUMEN

AR-12 has been evaluated in clinical trials as an anti-cancer agent but also has demonstrated host-directed, broad-spectrum clearance of bacteria. We have previously shown that AR-12 has activity in vitro against Salmonella enterica serovar Typhimurium and Francisella species by inducing autophagy and other host immune pathways. AR-12 treatment of S. Typhimurium-infected mice resulted in a 10-fold reduction in bacterial load in the liver and spleen and an increased survival time. However, AR-12 treatment did not protect mice from death, likely due poor formulation. In the current study, AR-12 was encapsulated in a microparticulate carrier formulated from the novel degradable biopolymer acetalated dextran (Ace-DEX) and subsequently evaluated for its activity in human monocyte-derived macrophages (hMDMs). Our results show that hMDMs efficiently internalized Ace-DEX microparticles (MPs), and that encapsulation significantly reduced host cell cytotoxicity compared to unencapsulated AR-12. Efficient macrophage internalization of AR-12 loaded MPs (AR-12/MPs) was further demonstrated by autophagosome formation that was comparable to free AR-12 and resulted in enhanced clearance of intracellular Salmonella. Taken together, these studies provide support that Ace-DEX encapsulated AR-12 may be a promising new therapeutic agent to control intracellular bacterial pathogens of macrophages by targeting delivery and reducing drug toxicity.


Asunto(s)
Antibacterianos/administración & dosificación , Dextranos/química , Portadores de Fármacos/química , Pirazoles/administración & dosificación , Salmonella typhimurium/efectos de los fármacos , Sulfonamidas/administración & dosificación , Acetales/química , Antibacterianos/farmacología , Autofagia/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Composición de Medicamentos , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Pirazoles/farmacología , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/microbiología , Sulfonamidas/farmacología , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA