Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Chin Med Assoc ; 86(1): 26-33, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36599139

RESUMEN

BACKGROUND: Receptor interacting serine/threonine kinase 1 (RIPK1) mediates apoptosis by regulating the classic proapoptotic effectors Bcl-2-associated X protein (Bax) and Bcl-2 homologous antagonist/killer (Bak). Although Bcl-2-related ovarian killer (Bok) is structurally similar to Bak and Bax, it is unclear whether it mediates apoptosis in skeletal muscle ischemia reperfusion (IR) injury. We hypothesized that by regulating Bok-mediated apoptosis, inhibiting RIPK1 with necrostatin-1 would reduce skeletal muscle IR injury. METHODS: Rats were randomized into four groups: sham (SM), IR, IR treated with necrostatin-1 (NI), or vehicle dimethyl sulfoxide (DI). For the IR group, the right femoral artery was clamped for 4 hours and then reperfused for 4 hours, and for the NI and DI groups, necrostatin-1 (1.65 mg/kg) and the equal volume of dimethyl sulfoxide were intraperitoneally administered prior to IR induction. The structural damage of muscle tissue and protein expression of Bok, Bcl-2, and cleaved caspase-3 were investigated, and apoptotic cells were identified with terminal dUTP nick-end labeling (TUNEL) staining. In vitro, human skeletal muscle cells (HSMCs) were exposed to 6 hours of oxygen-glucose deprivation followed by normoxia for 6 hours to establish an oxygen-glucose deprivation/reoxygenation (OGD/R) model. To determine the role of Bok, cell viability, lactate dehydrogenase (LDH) release, and flow cytometry were examined to demonstrate the effects of necrostatin-1 and Bok knockdown on the OGD/R insult of HSMCs. RESULTS: Necrostatin-1 pretreatment markedly reduced IR-induced muscle damage and RIPK1, Bok, and cleaved caspase-3 expression, whereas upregualted Bcl-2 expression (p < 0.05). Furthermore, necrostatin-1 prevented mitochondrial damage and decreased TUNEL-positive muscle cells (p < 0.05). In vitro, HSMCs treated with necrostatin-1 showed reduced Bok expression, increased cell viability, and reduced LDH release in response to OGD/R (p < 0.05), and Bok knockdown significantly blunted the OGD/R insult in HSMCs. CONCLUSION: Necrostatin-1 prevents skeletal muscle from IR injury by regulating Bok-mediated apoptosis.


Asunto(s)
Dimetilsulfóxido , Daño por Reperfusión , Ratas , Humanos , Animales , Proteína X Asociada a bcl-2 , Caspasa 3/metabolismo , Caspasa 3/farmacología , Dimetilsulfóxido/farmacología , Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2 , Daño por Reperfusión/prevención & control , Oxígeno , Músculo Esquelético/metabolismo , Glucosa
2.
Huan Jing Ke Xue ; 42(11): 5433-5439, 2021 Nov 08.
Artículo en Zh | MEDLINE | ID: mdl-34708982

RESUMEN

In this work, a novel sodium silicate-modified peanut shell biochar(Si-PSB) was synthesized and used as phosphorus adsorbents. Compared with unmodified biochar(PSB), the adsorption capacity of Si-PSBs increased significantly. The adsorption capacity of 8% sodium silicate solution modified biochar(8%Si-PSB) was 3.9 times higher than that of PSB. The biochar was characterized using scanning electron microscopy(SEM), Fourier transformed infrared(FTIR), and X-ray diffraction(XRD), which confirmed that silica was present on the surface of 8%Si-PSB. The introduction of silica improved the reaction activity of biochar's own metal ions by affecting the morphology of calcium carbonate. The 8%Si-PSB had a good adsorption effect on phosphorus in both acid and alkali environments. Phosphorus adsorption by 8%Si-PSB and PSB was described well by the pseudo-second-order model, and the adsorption capacity after equilibrium fluctuated between 2.79 mg·g-1 and 0.71 mg·g-1, respectively. Further, the isothermal adsorption experimental data fitted well to the Langmuir model. The presence of humic acid in the solution inhibited the adsorption of phosphorus by the 8%Si-PSB and PSB. The 8%Si-PSB, as a new low-cost phosphorus removal material, can improve the utilization of metal ions in peanut shell itself.


Asunto(s)
Fósforo , Contaminantes Químicos del Agua , Adsorción , Arachis , Carbón Orgánico , Cinética , Espectroscopía Infrarroja por Transformada de Fourier , Contaminantes Químicos del Agua/análisis
3.
Anesth Analg ; 111(2): 539-43, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20610550

RESUMEN

BACKGROUND: Acute lung injury is a recognized complication of lower limb ischemia-reperfusion that has been demonstrated experimentally and in the clinical setting of aortic surgery. The application of a tourniquet can cause lower limb ischemia-reperfusion in orthopedic surgery. We studied the effect of unilateral thigh tourniquet-induced lower limb ischemia-reperfusion on pulmonary function, and the role of ischemic preconditioning in attenuating pulmonary dysfunction. METHODS: Thirty ASA I or II patients scheduled for lower extremity surgery were randomized into 2 groups: a limb ischemia-reperfusion group with tourniquet application (ischemia-reperfusion group, n = 15) and an ischemia preconditioning group (preconditioning group, n = 15), in which patients received 3 cycles of 5 minutes of ischemia, alternating with 5 minutes of reperfusion before extended use of the tourniquet. Blood gas, plasma malondialdehyde, and serum interleukin-6 (IL-6), IL-8, and IL-10 levels were measured just before tourniquet inflation, 1 hour after inflation and 2 hours, 6 hours, and 24 hours after tourniquet deflation. Arterial-alveolar oxygen tension ratio, alveolar-arterial oxygen tension difference, and respiratory index also were calculated. RESULTS: In comparison with the baseline values, arterial Po(2) and arterial-alveolar oxygen tension ratio were decreased, while alveolar-arterial oxygen tension difference and respiratory index were increased significantly 6 hours after tourniquet deflation in both groups (P < 0.01). However, these changes were less significant in the ischemic preconditioning group than those in the lower limb ischemia-reperfusion group (P < 0.01). Similarly, the increases in the malondialdehyde, IL-6, and IL-8 from 2 hours to 24 hours after release of the tourniquet in the lower limb ischemia-reperfusion group were attenuated by ischemic preconditioning. CONCLUSIONS: Pulmonary gas exchange is impaired after lower limb ischemia-reperfusion associated with the clinical use of a tourniquet for lower limb surgery. Ischemic preconditioning preceding tourniquet-induced ischemia attenuates lipid peroxidation and systemic inflammatory response and mitigates pulmonary dysfunction.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Precondicionamiento Isquémico/métodos , Pulmón/fisiopatología , Procedimientos Ortopédicos/efectos adversos , Intercambio Gaseoso Pulmonar , Daño por Reperfusión/terapia , Muslo/irrigación sanguínea , Torniquetes/efectos adversos , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/fisiopatología , Adulto , Biomarcadores/sangre , Dióxido de Carbono/sangre , Distribución de Chi-Cuadrado , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-10/sangre , Interleucina-6/sangre , Interleucina-8/sangre , Peroxidación de Lípido , Pulmón/metabolismo , Masculino , Malondialdehído/sangre , Persona de Mediana Edad , Oxígeno/sangre , Estudios Prospectivos , Flujo Sanguíneo Regional , Daño por Reperfusión/sangre , Daño por Reperfusión/etiología , Daño por Reperfusión/fisiopatología , Factores de Tiempo , Resultado del Tratamiento
4.
Chin J Integr Med ; 23(1): 18-24, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27299461

RESUMEN

OBJECTIVE: To compare the effect between nebulized and intravenous administration of Shenmai Injection () on pulmonary gas exchange function of patients following tourniquet-induced lower limb ischemia-reperfusion. METHODS: Thirty-eight patients scheduled for lower extremity surgery were randomized into three groups using the closed envelop method: Shenmai Injection was administered 30 min before tourniquet inflflation by nebulization [0.6 mL/kg in 10 mL normal saline (NS)] in the nebulization group or by intravenous drip (0.6 mL/kg dissolved in 250 mL of 10% glucose) in the intravenous drip group, and equal volume of NS was given intravenously in the NS group; 15 in each group. Arterial blood gases were analyzed, serum levels of malonaldehyde (MDA) and interleukine-6 (IL-6) and interleukine-8 (IL-8) were determined using the method of thiobarbituric acid reaction and enzyme-linked immuno sorbent assay respectively just before tourniquet inflflation (T0), and at 0.5 h (T1), 2 h (T2), 6 h (T3) after tourniquet deflflation. RESULTS: Compared with baselines at T0, MDA levels signifificantly increased at T2, T3 in the NS group and at T3 in the nebulization group, and IL-6 and IL-8 levels were signifificantly increased at T2, T3 in NS, the intravenous drip and the nebulization groups (P <0.05). Arterial pressure of oxygen (PaO2) at T3 was decreased, while alveolararterial oxygen tension showed difference (PA-aDO2) at T3 in the NS group; RI at T3 in both intravenous drip and the nebulization groups were enhanced (P <0.05). Compared with the NS group, MDA and IL-8 levels at T2, T3, IL-6 at T3 in the intravenous drip group, and IL-8 at T3 in the nebulization group were all remarkably increased (P <0.05). Additionally, MDA level at T3 in the nebulization group was higher than that in the intravenous drip group (P <0.05). CONCLUSIONS: Intravenous administration of Shenmai Injection provided a better protective effect than nebulization in mitigating pulmonary gas exchange dysfunction in patients following tourniquet-induced limb ischemia-reperfusion.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/uso terapéutico , Intercambio Gaseoso Pulmonar , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/fisiopatología , Torniquetes/efectos adversos , Adulto , Análisis de los Gases de la Sangre , Vías de Administración de Medicamentos , Combinación de Medicamentos , Medicamentos Herbarios Chinos/farmacología , Femenino , Humanos , Inyecciones , Interleucina-6/sangre , Interleucina-8/sangre , Masculino , Malondialdehído/sangre , Intercambio Gaseoso Pulmonar/efectos de los fármacos , Daño por Reperfusión/sangre
5.
Int Immunopharmacol ; 24(2): 247-255, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25479721

RESUMEN

Roxithromycin (RXM) expresses anti-asthmatic effects that are separate from its antibiotic activity, but its effects on airway remodeling are still unknown. Here, we evaluated the effects of RXM on airway remodeling and the expression of caveolin-1 and phospho-p42/p44mitogen-activated protein kinase (phospho-p42/p44MAPK) in chronic asthmatic rats. The chronic asthma was induced by ovalbumin/Al(OH)3 sensitization and ovalbumin challenge, RXM (30mg/kg) or dexamethasone (0.5mg/kg) was given before airway challenge initiation. We measured the thickness of bronchial wall and bronchial smooth muscle cell layer to indicate airway remodeling, and caveolin-1 and phospho-p42/p44MAPK expression in lung tissue and airway smooth muscle were detected by immunohistochemistry and western blot analysis, respectively. The results demonstrated that RXM treatment decreased the thickness of bronchial wall and bronchial smooth muscle cell layer, and also downregulated the phospho-p42/p44MAPK expression and upregulated the caveolin-1 expression. The above effects of RXM were similar to dexamethasone. Our results suggested that pretreatment with RXM could suppress airway remodeling and regulate the expression of caveolin-1 and phospho-p42/p44MAPK in chronic asthmatic rats.


Asunto(s)
Asma/tratamiento farmacológico , Bronquios/efectos de los fármacos , Caveolina 1/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Roxitromicina/administración & dosificación , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Alérgenos/inmunología , Animales , Bronquios/patología , Caveolina 1/genética , Enfermedad Crónica , Dexametasona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Miocitos del Músculo Liso/fisiología , Ovalbúmina/inmunología , Ratas , Ratas Sprague-Dawley , Roxitromicina/farmacología
6.
Int J Nanomedicine ; 6: 1075-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23515704

RESUMEN

BACKGROUND: Mechanical ventilation has been documented to paradoxically cause lung injury. As a commonly used volatile anesthetic, sevoflurane has been proven to possess antiinflammatory and antioxidative properties. This study aims to investigate the protective effects of sevoflurane on inflammation and ventilator-induced lung injury during mechanical ventilation in healthy mice. METHODS: The adult healthy mice were divided into four groups, each consisting of ten subjects: mice in group Con-L(VT) and group Sev-L(VT) were ventilated with tidal volumes of 8 mL/kg for 4 hours, while those in group Con-H(VT) and group Sev-H(VT) were ventilated with tidal volumes of 16 mL/kg instead. Control mice (group Con-L(VT) and Con-H(VT)) were subjected to fresh air, while sevoflurane-treated mice (groups Sev- L(VT) and Sev-H(VT)) were subjected to air mixed with 1 vol% sevoflurane. After 4 hours of ventilation, the bronchoalveolar lavage (BAL) fluid was collected and analyzed for the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and IL-10. Lung homogenates were harvested to detect the expression of nuclear factor-kappa B (NF-κB) and heme oxygenase (HO)-1 mRNA by reverse transcription-polymerase chain reaction method. Lung damage was evaluated using the modified Ventilator-Induced Lung Injury histological scoring system. RESULTS: Compared to group Con-L(VT), the levels of TNF-α, IL-1ß, IL-6, and IL-10 in BAL fluid, mRNA expressions of NF-κB and HO-1 in lung tissue, and lung injury scores were significantly increased in group Con-H(VT); compared to group Con-H(VT), group Sev-H(VT) BAL samples showed decreased levels of TNF-α, IL-1ß, and IL-6; they also showed increased levels of IL-10, the downregulation of NF-κB mRNA, and HO-1 mRNA upregulation; the lung injury scores were significantly lower in group Sev-H(VT) than group Con-H(VT). CONCLUSION: Mechanical ventilation with high tidal volume might lead to lung injury, which could be significantly, but not completely, attenuated by sevoflurane inhalation by inhibiting the NF-κB-mediated proinflammatory cytokine generation and upregulating HO-1 expression.


Asunto(s)
Antiinflamatorios/farmacología , Hemo-Oxigenasa 1/genética , Proteínas de la Membrana/genética , Éteres Metílicos/farmacología , Neumonía/tratamiento farmacológico , ARN Mensajero/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Análisis de Varianza , Animales , Líquido del Lavado Bronquioalveolar/química , Citocinas/análisis , Citocinas/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hemodinámica , Pulmón/química , Pulmón/metabolismo , Pulmón/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neumonía/genética , Neumonía/metabolismo , ARN Mensajero/genética , Sevoflurano , Estadísticas no Paramétricas , Regulación hacia Arriba/efectos de los fármacos , Lesión Pulmonar Inducida por Ventilación Mecánica/genética , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo
7.
Int Immunopharmacol ; 16(2): 288-95, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23587488

RESUMEN

BACKGROUND: Mechanical ventilation especially with large tidal volume has been demonstrated to activate inflammatory response inducing lung injury, which could be attenuated by cyclooxygenase (COX)-2 inhibitors. As the main small integral membrane proteins that selectively conduct water molecules' transportation, aquaporin (AQP)-1 downregulation significantly related to lung edema and inflammation. This study aims to investigate the role of AQP1 in ventilator-induced lung injury in rats and evaluates the effects of COX-2 inhibition. METHODS: Forty rats were allocated into four groups, where rats in Groups LD (low volume+DMSO) and LN (low volume+NS-398) were given intravenously 2ml DMSO and 8mg/kg NS-398 (a specific COX-2 inhibitor, dissolved in 2ml DMSO) before 4-hour lower tidal volume ventilation (8ml/kg), respectively, while DMSO and NS-398 were administrated in the same manner before 4-hour injurious ventilation (40ml/kg) in Groups HD (high volume+DMSO) and HN (high volume+NS-398). The arachidonic acid metabolites (6-keto prostaglandin F1α, thromboxane B2), inflammatory cytokines (tumor necrosis factor-α, interleukin-1ß, 6, 8) and total protein levels in bronchoalveolar lavage (BAL) fluid and COX-2 mRNA and AQP1 protein expression in lung tissue were detected; water content and lung morphology were also evaluated. RESULTS: Compared to Groups LD and LN, the rats in Groups HD and HN suffered obvious lung morphological changes with higher wet-to-dry weight ratio and lung injury score, and the levels of arachidonic acid metabolites, inflammatory cytokines and total protein in BAL fluid were increased, the expression of COX-2 mRNA was significantly upregulated and AQP1 protein was downregulated in lung tissue (p<0.05). The changes in BAL fluid and the severity of lung injury were attenuated, and AQP1 expression was upregulated in Group HN as compared to HD (p<0.05). CONCLUSIONS: Ventilation with large tidal volume causes inflammatory mediator production and AQP1 downregulation, which could be attenuated by COX-2 inhibition.


Asunto(s)
Acuaporina 1/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Lesión Pulmonar/metabolismo , Nitrobencenos/farmacología , Respiración Artificial/efectos adversos , Sulfonamidas/farmacología , Animales , Líquido del Lavado Bronquioalveolar/química , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Citocinas/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Nitrobencenos/uso terapéutico , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Sulfonamidas/uso terapéutico
8.
Int Immunopharmacol ; 13(3): 280-3, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22579844

RESUMEN

BACKGROUND: Mechanical ventilation may paradoxically cause lung injury. Protective mechanical ventilation strategy utilizing low tidal volume and high frequency has been shown to attenuate inflammation and reduce mortality in non-diabetic patients. The purpose of this present study was to observe the effects of diabetes on inflammation and lung injury in mice with protective ventilation strategy. METHODS: Forty mice were included in our study. The mice in Group Dia-MV and Con-MV were subjected to 4 hour-ventilation. And the mice in Group Dia-SB and Con-SB were exposed to room air breathing spontaneously for 4h. Tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-10 (IL-10), superoxide dismutase (SOD) and malondialdehyde (MDA) levels in serum were detected and the expression of inflammatory cytokine mRNA was also determined in lung tissue. Lung damage was assessed using a modified lung injury score. RESULTS: The serum levels of TNF-α, IL-6, and IL-10 in Group Dia-MV were significantly higher than those in Group Dia-SB or Group Con-MV or Group Con-SB (P<0.05). Quantitative RT-PCR analysis of pro-inflammatory cytokines in lung homogenates presented similar results. The mice in Group Dia-MV suffered obvious lung histological changes, whose lung injury scores were significantly higher in Group Dia-SB as compared to Group Con-SB , Group Con-MV or Group Dia-SB (P<0.05). CONCLUSIONS: Diabetes increased the inflammation reaction and associated lung injury in mice in spite of the protective mechanical ventilation strategy based on low tidal volumes and high frequency.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/fisiopatología , Inflamación/etiología , Lesión Pulmonar/etiología , Respiración Artificial/efectos adversos , Animales , Citocinas/biosíntesis , Citocinas/sangre , Citocinas/genética , Diabetes Mellitus Experimental/complicaciones , Inflamación/inmunología , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/inmunología , Lesión Pulmonar/patología , Lesión Pulmonar/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Respiración Artificial/métodos , Volumen de Ventilación Pulmonar , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Chin Med J (Engl) ; 124(3): 457-60, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21362350

RESUMEN

BACKGROUND: Aquaporin-1 (AQP1) has involved in fluid transport in diverse pulmonary edema diseases. Our study aimed to explore the dynamic changes of AQP1 in pulmonary water metabolism in rats following traumatic brain injury (TBI) and the protective effect provided by shenmai injection. METHODS: Sixty male Sprague Dawley rats weighting 280 - 300 g were randomly divided into three groups: the normal control group, the model group and the shenmai injection (SMI) group. One piece skull was taken away without injuring cerebral tissue in normal control group, while rats in model group and SMI group were subject to free fall injury in the cerebral hemisphere. Rats in model group received intraperitoneal normal sodium (15 ml/kg) at one hour post-injury and the same dose of shenmai injection instead in SMI group, respectively. The expression of AQP1 was detected by immunohistochemical analysis and semi-quantitative RT-PCR at 0 hour, 10 hours, 72 hours and 120 hours after TBI. Arterial blood gas analysis and lung wet to dry were also measured. RESULTS: AQP1 was mainly presented in the capillary endothelium and slightly alveolar epithelial cells in three groups, but the expression of AQP1 in the normal control group was positive and tenuous, weakly positive in the model and SMI groups, respectively. Compared with normal control group, AQP1 mRNA levels were down regulated in the model and SMI groups at 10 hours, 72 hours and 120 hours (P < 0.05). While AQP1 mRNA levels in the SMI group was up-regulated than that in the model group (P < 0.05). Lung wet to dry weight ratio (W/D) in the model and SMI groups at 10 hours were higher than that in normal control group (P < 0.05). Compared with normal control group, PaO2 was markedly lower in the model and SMI groups (P < 0.05), but there were no statistically significant differences between model and SMI groups (P > 0.05). CONCLUSIONS: The decreased AQP1 expression may be involved in the increased lung water content and dysfunction of pulmonary water metabolism following TBI. The treatment with SMI could improve water metabolism by promoting AQP1 expression.


Asunto(s)
Acuaporina 1/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Medicamentos Herbarios Chinos/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Animales , Acuaporina 1/genética , Combinación de Medicamentos , Medicamentos Herbarios Chinos/administración & dosificación , Inmunohistoquímica , Inyecciones , Masculino , Distribución Aleatoria , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA