Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Pregnancy Childbirth ; 22(1): 221, 2022 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-35305610

RESUMEN

AIM: To establish a model for predicting adverse outcomes in advanced-age pregnant women with preterm preeclampsia in China. METHODS: We retrospectively collected the medical records of 896 pregnant women with preterm preeclampsia who were older than 35 years and delivered at the Affiliated Hospital of Qingdao University from June 2018 to December 2020. The pregnant women were divided into an adverse outcome group and a non-adverse outcome group according to the occurrence of adverse outcomes. The data were divided into a training set and a verification set at a ratio of 8:2. A nomogram model was developed according to a binary logistic regression model created to predict the adverse outcomes in advanced-age pregnant women with preterm preeclampsia. ROC curves and their AUCs were used to evaluate the predictive ability of the model. The model was internally verified by using 1000 bootstrap samples, and a calibration diagram was drawn. RESULTS: Binary logistic regression analysis showed that platelet count (PLT), uric acid (UA), blood urea nitrogen (BUN), prothrombin time (PT), and lactate dehydrogenase (LDH) were the factors that independently influenced adverse outcomes (P < 0.05). The AUCs of the internal and external verification of the model were 0.788 (95% CI: 0.737 ~ 0.764) and 0.742 (95% CI: 0.565 ~ 0.847), respectively. The calibration curve was close to the diagonal. CONCLUSIONS: The model we constructed can accurately predict the risk of adverse outcomes of pregnant women of advanced age with preterm preeclampsia, providing corresponding guidance and serving as a basis for preventing adverse outcomes and improving clinical treatment and maternal and infant prognosis.


Asunto(s)
Edad Materna , Nomogramas , Preeclampsia/patología , Complicaciones del Embarazo/epidemiología , Resultado del Embarazo/epidemiología , Adulto , Pueblo Asiatico/etnología , China/epidemiología , Femenino , Humanos , Embarazo , Embarazo de Alto Riesgo/etnología , Pronóstico , Curva ROC , Estudios Retrospectivos , Factores de Riesgo , Sensibilidad y Especificidad
2.
Mol Ther ; 28(1): 313-327, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31604679

RESUMEN

MicroRNAs (miRNAs) play an essential role in the self-renewal of breast cancer stem cells (BCCs). Our study aimed to clarify the role of proto-oncogene c-Jun-regulated miR-5188 in breast cancer progression and its association with Timeless-mediated cancer stemness. In the present study, we showed that miR-5188 exerted an oncogenic effect by inducing breast cancer stemness, proliferation, metastasis, and chemoresistance in vitro and in vivo. The mechanistic analysis demonstrated that miR-5188 directly targeted FOXO1, which interacted with ß-catenin in the cytoplasm, facilitated ß-catenin degradation, and impaired the nuclear accumulation of ß-catenin, thus stimulating the activation of known Wnt targets, epithelial-mesenchymal transition (EMT) markers, and key regulators of cancer stemness. Moreover, miR-5188 potentiated Wnt/ß-catenin/c-Jun signaling to promote breast cancer progression. Interestingly, c-Jun enhanced miR-5188 transcription to form a positive regulatory loop, and Timeless interacted with Sp1/c-Jun to induce miR-5188 expression by promoting c-Jun-mediated transcription, which further activated miR-5188-FOXO1/ß-catenin-c-Jun loop and facilitated breast cancer progression. Importantly, miR-5188 was upregulated in breast cancer and was positively correlated with poor patient prognosis. This study identifies miR-5188 as a novel oncomiR and provides a new theoretical basis for the clinical use of miR-5188 antagonists in the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína Forkhead Box O1/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ubiquitinación/genética , beta Catenina/metabolismo , Adulto , Animales , Neoplasias de la Mama/patología , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Pronóstico , Proto-Oncogenes Mas , Transfección , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
3.
BMC Pregnancy Childbirth ; 21(1): 581, 2021 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-34420518

RESUMEN

AIM: To establish a nomogram model to predict the risk of macrosomia in pregnant women with gestational diabetes mellitus in China. METHODS: We retrospectively collected the medical records of 783 pregnant women with gestational diabetes who underwent prenatal examinations and delivered at the Affiliated Hospital of Qingdao University from October 2019 to October 2020. The pregnant women were randomly divided into two groups in a 4:1 ratio to generate and validate the model. The independent risk factors for macrosomia in pregnant women with gestational diabetes mellitus were analyzed by multivariate logistic regression, and the nomogram model to predict the risk of macrosomia in pregnant women with gestational diabetes mellitus was established and verified by R software. RESULTS: Logistic regression analysis showed that prepregnancy body mass index, weight gain during pregnancy, fasting plasma glucose, triglycerides, biparietal diameter and amniotic fluid index were independent risk factors for macrosomia (P < 0.05). The areas under the ROC curve for internal and external validation of the model were 0.813 (95 % confidence interval 0.754-0.862) and 0.903 (95 % confidence interval 0.588-0.967), respectively. The calibration curve was a straight line with a slope close to 1. CONCLUSIONS: In this study, we constructed a nomogram model to predict the risk of macrosomia in pregnant women with gestational diabetes mellitus. The model has good discrimination and calibration abilities, which can help clinical healthcare staff accurately predict macrosomia in pregnant women with gestational diabetes mellitus.


Asunto(s)
Diabetes Gestacional/epidemiología , Macrosomía Fetal/complicaciones , Macrosomía Fetal/epidemiología , Nomogramas , Medición de Riesgo/métodos , Adulto , China/epidemiología , Femenino , Humanos , Embarazo , Reproducibilidad de los Resultados , Estudios Retrospectivos , Factores de Riesgo , Adulto Joven
4.
Breast Cancer Res ; 19(1): 53, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28464854

RESUMEN

BACKGROUND: Breast cancer is the most common malignancy and the leading cause of cancer death among women. TIMELESS (TIM), a circadian rhythm regulator, has been recently implicated in the progression of human cancer. However, the role of TIM in the progression of breast cancer has not been well-characterized. METHODS: Immunohistochemistry (IHC) staining was used to examine TIM levels in breast cancer specimens. Mammosphere formation analysis and side population analysis were used to examine the effect of TIM on the self-renewal of breast cancer stem cells. A wound healing assay and a Transwell assay were used to determine the role of TIM in breast cancer cell migration and invasion. A soft agar growth assay in vitro and tumorigenicity in vivo were used to determine the role of TIM in tumorigenicity. RESULTS: TIM levels in both breast cancer cell lines and tissues were significantly upregulated. Patients with high TIM had poorer prognosis than patients with low TIM. Overexpression of TIM dramatically enhanced, while knockdown of TIM suppressed the self-renewal of cancer stem cells (CSCs), cell invasion and migration abilities of breast cancer cells in vitro. Moreover, overexpression of TIM significantly augmented, while knockdown of TIM reduced the tumorigenicity of breast cancer cells in vivo. Mechanism studies revealed that TIM upregulated the expression and the trans-activity of the well-known oncogene MYC. Inhibition of MYC significantly blocked the effects of TIM on CSC population, cell invasion and anchor-independent cell growth. CONCLUSION: TIM plays an important role in promoting breast cancer progression and may represent a novel therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Proto-Oncogénicas c-myc/genética , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Invasividad Neoplásica/genética , Células Madre Neoplásicas/patología
5.
Acta Pharmacol Sin ; 38(2): 241-251, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27890917

RESUMEN

Sulforaphane is a common antioxidant selectively abundant in cruciferous plants, which exhibits effective anti-cancer actions in control of tumorigenesis or progression of various cancers. A recent study has shown that sulforaphane attenuates the EGFR signaling pathway in non-small cell lung cancer (NSCLC), suggesting its potential anti-metastatic effects. In this study we assessed the involvement of sulforaphane and miR-616-5p in epithelial-mesenchymal transition (EMT) and NSCLC metastasis. Sulforaphane suppressed the cell proliferation in human NSCLC cell lines H1299, 95C and 95D with IC50 values of 9.52±1.23, 9.04±1.90 and 17.35±2.03 µmol/L, respectively. At low concentrations (1-5 µmol/L), sulforaphane dose-dependently inhibited the migration and invasion of 95D and H1299 cells with relatively high metastatic potential. The anti-metastatic action of sulforaphane was confirmed in 95D and H1299 cell xenografts in vivo. In fresh NSCLC tissue samples from 179 patients, miR-616-5p levels were upregulated in late-stage NSCLCs, and strongly correlated with risk of NSCLC recurrence and metastasis. Consistent with the clinic observation, miR-616-5p levels in the 3 NSCLC cell lines were correlated with their metastatic ability, and were decreased by sulforaphane treatment. Silencing miR-616-5p markedly suppressed the migration and invasion of 95D cells in vitro and NSCLC metastasis in vivo. Further studies revealed that miR-616-5p directly targeted GSK3ß and decreased its expression, whereas sulforaphane decreased miR-616-5p levels by histone modification, and followed by inactivation of the GSK3ß/ß-catenin signaling pathway and inhibition of EMT, which was characterized by loss of epithelial markers and acquisition of a mesenchymal phenotype in NSCLC cells. Our findings suggest that sulforaphane is a potential adjuvant chemotherapeutic agent for the prevention of NSCLC recurrence and metastasis, and miR-616-5p can be clinically utilized as a biomarker or therapeutic target to inhibit metastasis.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Isotiocianatos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , MicroARNs/metabolismo , Metástasis de la Neoplasia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Isotiocianatos/farmacología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Sulfóxidos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Nanosci Nanotechnol ; 12(6): 4825-31, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22905536

RESUMEN

Many of today's demanding applications require thin-film coatings with high hardness, toughness, and thermal stability. In many cases, coating thickness in the range 2-20 microm and low surface roughness are required. Diamond films meet many of the stated requirements, but their crystalline nature leads to a high surface roughness. Nanocrystalline diamond offers a smoother surface, but significant surface modification of the substrate is necessary for successful nanocrystalline diamond deposition and adhesion. A hybrid hard and tough material may be required for either the desired applications, or as a basis for nanocrystalline diamond film growth. One possibility is a composite system based on carbides or nitrides. Many binary carbides and nitrides offer one or more mentioned properties. By combining these binary compounds in a ternary or quaternary nanocrystalline system, we can tailor the material for a desired combination of properties. Here, we describe the results on the structural and mechanical properties of the coating systems composed of tungsten-chromium-carbide and/or nitride. These WC-Cr-(N) coatings are deposited using magnetron sputtering. The growth of adherent nanocrystalline diamond films by microwave plasma chemical vapor deposition has been demonstrated on these coatings. The WC-Cr-(N) and WC-Cr-(N)-NCD coatings are characterized with atomic force microscopy and SEM, X-ray diffraction, X-ray photoelectron spectroscopy, Raman spectroscopy, and nanoindentation.


Asunto(s)
Cristalización/métodos , Diamante/química , Nanoestructuras/química , Nanoestructuras/ultraestructura , Tungsteno/química , Calor , Sustancias Macromoleculares/química , Ensayo de Materiales , Conformación Molecular , Tamaño de la Partícula , Propiedades de Superficie
7.
Front Genet ; 13: 991936, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36118852

RESUMEN

Purpose: Hepatocellular carcinoma (HCC) is a severe malignant tumor with high incidence and mortality. LncRNAs present broad clinical application prospects. Herein, we aim to identify a cancer stemness associated lncRNA and reveal its role in HCC diagnosis, prognosis evaluation, and progression. Methods: The cancer stemness-associated LINC02475 in HCC samples were identified using bioinformatics analysis. Cellular and molecular experiments were conducted to elucidate the role of LINC02475 in HCC. Results: The firm links between LINC02475 and HCC stemness and prognosis were demonstrated by bioinformatics analysis of public cancer datasets. LINC02475 expression was elevated in HCC, performed well in the diagnosis, and independently predicts poor overall survival (hazard ratio = 1.389, 95% confidence interval = 1.047-1.843, p = 0.023), as well as progression-free survival (hazard ratio = 1.396, 95% confidence interval = 1.016-1.917, p = 0.040) of HCC patients. Moreover, LINC02475 enhanced the tumorigenic pathways necessary for cell stemness, DNA replication required for cell proliferation, epithelial-mesenchymal transition involved in metastasis, and DNA damage repair pathways that drove cell radioresistance and cisplatin resistance, thus promoting HCC progression. Conclusion: Cancer stemness-associated LINC02475 independently predicted a poor prognosis and promoted HCC progression by enhancing stemness, proliferation, metastasis, radioresistance, and chemoresistance. Our study lays a foundation for the clinical application of LINC02475 as a novel biomarker and target for the diagnosis, prognosis evaluation, as well as treatment of HCC.

8.
Phytomedicine ; 105: 154364, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35932608

RESUMEN

BACKGROUND: Developing novel and effective drugs with less toxicity is urgent for non-small cell lung cancer (NSCLC) therapy. Xanthotoxol (Xan) is the major natural component of the medical plant Angelica dahurica with potential anti-cancer activities. PURPOSE: In this study, we aimed to demonstrate the effect and underlying mechanism of Xan in NSCLC and evaluate the effectiveness of Xan in NSCLC patients. METHODS: CCK8, colony formation, EdU, flow cytometry, and transwell assays were carried out to investigate the anti-NSCLC activity of Xan in vitro. In addition, the xenograft mouse model was established to evaluate the anti-NSCLC effect of Xan in vivo. Moreover, bioinformatics analysis was performed to establish a prediction model based on RNA sequencing data. Furthermore, Western blot was used to detect the expression of proteins regulated by Xan. RESULTS: Xan inhibited the cell viability, colony formation capacity, DNA replication, cell cycle transition, migration and invasion, as well as inducing apoptosis of NSCLC cells. In addition, Xan suppressed NSCLC xenograft growth in vivo without obvious toxicity. Interestingly, bioinformatics analyses based on the RNA sequencing data indicated that Xan exerted inhibitory effects on NSCLC cells by down-regulating signals contributing to NSCLC progression and demonstrated that Xan was effective in ameliorating the prognosis of NSCLC patients with a new proposed prediction model. Moreover, Xan was shown to regulate cell cycle arrest, apoptosis, and epithelial-mesenchymal transition (EMT)-associated genes through downregulating PI3K-AKT signaling, thus suppressing NSCLC proliferation and metastasis. CONCLUSIONS: Taken together, our work proved that Xan induced cell cycle arrest, facilitated apoptosis, and inhibited EMT processes through downregulating the PI3K-AKT pathway to suppress NSCLC progress. Moreover, we also proposed a new model for evaluating Xan as a novel and effective drug in NSCLC treatments.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Furocumarinas , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Fosfatidilinositol 3-Quinasas , Pronóstico , Proteínas Proto-Oncogénicas c-akt
9.
Cell Death Discov ; 8(1): 497, 2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36566230

RESUMEN

N6-methylation of adenosine (m6A) is one of the most frequent chemical modifications in eukaryotic RNAs and plays a vital role in tumorigenesis and progression. Recently, emerging studies have shown that m6A modification by ALKBH5 was associated with immunotherapy response in various types of cancer. However, whether m6A demethylases ALKBH5 participate in regulating the tumor immune microenvironment and the efficacy of immunotherapy in glioblastoma remain unknown. Here, we found that deletion of ALKBH5 significantly inhibited the growth of glioma allografts, rescued the antitumoral immune response, and increased cytotoxic lymphocyte infiltration and proinflammatory cytokines in CSF while significantly suppressing PD-L1 protein expression. m6A-methylated RNA immunoprecipitation sequencing and RNA sequencing identify ZDDHC3 as the direct target of ALKBH5. Mechanically, ALKBH5 deficiency impairs the YTHDF2-mediated stability of ZDHHC3 mRNA, thereby suppressing PD-L1 expression by accelerating PD-L1 degradation in glioma. In addition, genetic deletion or pharmacological inhibition of ALKBH5 with IOX1 enhances the therapeutic efficacy of anti-PD-1 treatment in preclinical mice models. These data suggest that the combination of anti-PD-1 therapy and ALKBH5 inhibition may be a promising treatment strategy in glioma.

10.
Signal Transduct Target Ther ; 5(1): 13, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32296025

RESUMEN

MYH9 has dual functions in tumors. However, its role in inducing tumor stemness in hepatocellular carcinoma (HCC) is not yet determined. Here, we found that MYH9 is an effective promoter of tumor stemness that facilitates hepatocellular carcinoma pathogenesis. Importantly, targeting MYH9 remarkably improved the survival of hepatocellular carcinoma-bearing mice and promoted sorafenib sensitivity of hepatocellular carcinoma cells in vivo. Mechanistic analysis suggested that MYH9 interacted with GSK3ß and reduced its protein expression by ubiquitin-mediated degradation, which therefore dysregulated the ß-catenin destruction complex and induced the downstream tumor stemness phenotype, epithelial-mesenchymal transition, and c-Jun signaling in HCC. C-Jun transcriptionally stimulated MYH9 expression and formed an MYH9/GSK3ß/ß-catenin/c-Jun feedback loop. X protein is a hepatitis B virus (HBV)-encoded key oncogenic protein that promotes HCC pathogenesis. Interestingly, we observed that HBV X protein (HBX) interacted with MYH9 and induced its expression by modulating GSK3ß/ß-catenin/c-Jun signaling. Targeting MYH9 blocked HBX-induced GSK3ß ubiquitination to activate the ß-catenin destruction complex and suppressed cancer stemness and EMT. Based on TCGA database analysis, MYH9 was found to be elevated and conferred poor prognosis for hepatocellular carcinoma patients. In clinical samples, high MYH9 expression levels predicted poor prognosis of hepatocellular carcinoma patients. These findings identify the suppression of MYH9 as an alternative approach for the effective eradication of CSC properties to inhibit cancer migration, invasion, growth, and sorafenib resistance in HCC patients. Our study demonstrated that MYH9 is a crucial therapeutic target in HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/genética , Neoplasias Hepáticas/tratamiento farmacológico , Cadenas Pesadas de Miosina/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Xenoinjertos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Sorafenib/farmacología , Transactivadores/genética , Ubiquitinación/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/genética , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/genética
11.
Theranostics ; 9(25): 7583-7598, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31695788

RESUMEN

Cancer stem cells (CSCs) are the key factor in determining cancer recurrence, metastasis, chemoresistance and patient prognosis in hepatocellular carcinoma (HCC). The role of miR-5188 in cancer stemness has never been documented. In this study, we investigated the clinical and biological roles of miR-5188 in HCC. Methods: MiRNA expression in HCC was analyzed by bioinformatics analysis and in situ hybridization. The biological effect of miR-5188 was demonstrated in both in vitro and in vivo studies through the ectopic expression of miR-5188. The target gene and molecular pathway of miR-5188 were characterized using bioinformatics tools, dual-luciferase reporter assays, gene knockdown, and rescue experiments. Results: MiR-5188 was shown to be upregulated and confer poor prognosis in HCC patient data from TCGA database. MiR-5188 was subsequently identified as a significant inducer of cancer stemness that promotes HCC pathogenesis. Specifically, the targeting of miR-5188 by its antagomir markedly prolonged the survival time of HCC-bearing mice and improved HCC cell chemosensitivity in vivo. Mechanistic analysis indicated that miR-5188 directly targets FOXO1, which interacts with ß-catenin in the cytoplasm to reduce the nuclear translocation of ß-catenin and promotes the activation of Wnt signaling and downstream tumor stemness, EMT, and c-Jun. Moreover, c-Jun transcriptionally activates miR-5188 expression, forming a positive feedback loop. Interestingly, the miR-5188-FOXO1/ß-catenin-c-Jun feedback loop was induced by hepatitis X protein (HBX) through Wnt signaling and participated in the HBX-induced pathogenesis of HCC. Finally, analyses of transcriptomics data and our clinical data supported the significance of the abnormal expression of the miR-5188 pathway in HCC pathogenesis. Conclusions: These findings present the inhibition of miR-5188 as a novel strategy for the efficient elimination of CSCs to prevent tumor metastasis, recurrence and chemoresistance in patients with hepatocellular carcinoma. Our study highlights the importance of miR-5188 as a tumor stemness inducer that acts as a potential target for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Proteína Forkhead Box O1/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/fisiología , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Recurrencia Local de Neoplasia/metabolismo , Vía de Señalización Wnt/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA