Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Br J Anaesth ; 113 Suppl 1: i32-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24946779

RESUMEN

BACKGROUND: Lidocaine demethylates deoxyribonucleic acid (DNA) in breast cancer cells. This modification of epigenetic information may be of therapeutic relevance in the perioperative period, because a decrease in methylation can reactivate tumour suppressor genes and inhibit tumour growth. The objectives of this study were to determine the effect of two amide local anaesthetics, ropivacaine and bupivacaine, on methylation in two breast cancer cell lines and to detect whether the combination of lidocaine with the chemotherapy agent 5-aza-2'-deoxycytidine (DAC) would result in additive demethylating effects. METHODS: Breast cancer cell lines BT-20 [oestrogen receptor (ER)-negative] and MCF-7 (ER-positive) were incubated with lidocaine, bupivacaine, and ropivacaine to assess demethylating properties. Then, we tested varying concentrations of lidocaine and DAC to assess whether their demethylating effects were additive. Cell numbers and global methylation status were analysed. RESULTS: Lidocaine decreased methylation in BT-20 and MCF-7 cells, ropivacaine decreased methylation in BT-20 cells, and bupivacaine had no demethylating effect. When combined, lidocaine and DAC had additive demethylating effects. CONCLUSIONS: At clinically relevant doses, lidocaine and ropivacaine exert demethylating effects on specific breast cancer cell lines, but bupivacaine does not. The demethylating effects of lidocaine and DAC are indeed additive.


Asunto(s)
Amidas/farmacología , Anestésicos Locales/farmacología , Neoplasias de la Mama/genética , Metilación de ADN/efectos de los fármacos , Lidocaína/farmacología , Amidas/administración & dosificación , Anestésicos Locales/administración & dosificación , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias de la Mama/patología , Bupivacaína/administración & dosificación , Bupivacaína/farmacología , Supervivencia Celular/efectos de los fármacos , ADN de Neoplasias/genética , Decitabina , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Femenino , Humanos , Lidocaína/administración & dosificación , Células MCF-7 , Ropivacaína , Células Tumorales Cultivadas
2.
Br J Cancer ; 106(1): 189-98, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22108517

RESUMEN

BACKGROUND: The role of CHAC1 (cation transport regulator-like protein 1), a recently identified component of the unfolded protein response (UPR) pathway, in gynaecological cancers has not yet been characterised. Now, this work illustrates CHAC1 mRNA expression and associated clinical outcome in breast and ovarian cancer. METHODS: The prognostic value of CHAC1 and its two transcript variants was investigated in 116 breast and 133 ovarian tissues using quantitative real-time reverse-transcriptase PCR. Subsequently, we conducted functional studies using short-interfering RNA-mediated knockdown and plasmid-mediated overexpression of CHAC1 in breast and ovarian cancer cells. RESULTS: Poorly differentiated tumours exhibited higher CHAC1 mRNA expression (breast cancer: P=0.004; ovarian cancer: P=0.024). Hormone receptor-negative breast tumours and advanced-staged ovarian cancers demonstrated elevated CHAC1 mRNA expression levels (P<0.001 and P=0.026, respectively). The multivariate survival analysis showed a prognostic value of both transcript variants in breast cancer (transcript variant 1: RR(death) 6.7 (2.4-18.9); P<0.001), RR(relapse) 6.7 (2.1-21.3); P=0.001); (transcript variant 2: RR(death) 4.9 (2.0-12.4); P<0.001), RR(relapse) 8.0 (2.4-26.8); P<0.001). Ovarian cancer patients aged younger than 62.6 years with high CHAC1 mRNA expression showed poorer relapse-free- and overall-survival (P=0.030 and P=0.012, respectively). In functional studies CHAC1 knockdown suppressed cell migration, whereas ectopic overexpression opposed these effects. CONCLUSION: High CHAC1 mRNA expression could be an independent indicator for elevated risk of cancer recurrence in breast and ovarian cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Transporte de Catión/genética , Neoplasias Ováricas/patología , Empalme del ARN , ARN Mensajero/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Western Blotting , Neoplasias de la Mama/genética , Línea Celular Tumoral , Cartilla de ADN , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/genética
3.
Br J Anaesth ; 109(2): 200-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22542536

RESUMEN

BACKGROUND: Anaesthetic management of cancer surgery may influence tumour recurrence. The modulation of gene expression by methylation of deoxyribonucleic acid (DNA) (epigenetics) is increasingly recognized as a major hallmark of cancer. Next to direct effects of local anaesthetics upon tumour cells, the ester-type local anaesthetic, procaine, has been shown to affect methylation status in several tumour cell lines, promoting the reactivation of tumour suppressor genes. We sought to determine whether the prototype amide-type local anaesthetic, lidocaine, influences the survival and epigenetic status of oestrogen receptor (ER)-positive and -negative breast cancer cell lines in vitro. METHODS: Breast cancer cell lines BT-20 (ER-negative) and MCF-7 (ER-positive) were incubated with lidocaine and procaine as the reference substance. We performed cell count and determined apoptosis using TUNEL stain. Further, we assessed global methylation status, and methylation of three known tumour suppressor genes (RASSF1A, MYOD1, and GSTP1) using the MethyLight assay and real-time quantitative polymerase chain reaction, respectively. RESULTS: Baseline methylation was 100-fold higher in BT-20 cells. Here, we observed a dose-dependent decrease in DNA methylation in response to lidocaine (1, 0.01, and 0.01 mM) after 72 h (P<0.001, <0.001, and 0.004, respectively). The corresponding changes were smaller in MCF-7 cells. Global methylation status was profoundly influenced, but the methylation and mRNA expression status of three tumour suppressor genes was unchanged. CONCLUSIONS: Our findings suggest that demethylating tumour-suppressive effects of anaesthetic interventions may only be detectable in specific types of cancer due to differential methylation profiles. In conclusion, at clinically relevant concentrations, lidocaine demethylates DNA of breast cancer cell lines in vitro.


Asunto(s)
Anestésicos Locales/farmacología , Neoplasias de la Mama/genética , Metilación de ADN/efectos de los fármacos , Lidocaína/farmacología , Anestésicos Locales/administración & dosificación , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Lidocaína/administración & dosificación , Procaína/administración & dosificación , Procaína/farmacología , ARN Mensajero/genética , ARN Neoplásico/genética , Receptores de Estrógenos/metabolismo
4.
Neoplasma ; 59(6): 747-55, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22862176

RESUMEN

Because of its semi-solid character in dissemination and growth, advanced ovarian cancer with its hundreds of peritoneal tumor nodules and plaques appears to be an excellent in vivo model for studying the cancer stem cell hypothesis. The most important obstacle, however, is to adequately define and isolate these tumor-initiating cells endowed with the properties of anoikis-resistance and unlimited self-renewal. Until now, no universal single marker or marker constellation has been found to faithfully isolate (ovarian) cancer stem cells. As these multipotent cells are known to possess highly elaborated efflux systems for cytotoxic agents, these pump systems have been exploited to outline putative stem cells as a side-population (SP) via dye exclusion analysis. Furthermore, the cells in question have been isolated via flow cytometry on the basis of cell surface markers thought to be characteristic for stem cells.In the Vienna variant of the ovarian cancer cell line A2780 a proof-of-principle model with both a stable SP and a stable ALDH1A1+ cell population was established. Double staining clearly revealed that both cell fractions were not identical. Of note, A2780V cells were negative for expression of surface markers CD44 and CD117 (c-kit). When cultured on monolayers of healthy human mesothelial cells, green-fluorescence-protein (GFP)-transfected SP of A2780V exhibited spheroid-formation, whereas non-side-population (NSP) developed a spare monolayer growing over the healthy mesothelium. Furthermore, A2780V SP was found to be partially resistant to platinum. However, this resistance could not be explained by over-expression of the "excision repair cross-complementation group 1" (ERCC1) gene, which is essentially involved in the repair of platinated DNA damage. ERCC1 was, nonetheless, over-expressed in A2780V cells grown as spheres under stem cell-selective conditions as compared to adherent monolayers cultured under differentiating conditions. The same was true for the primary ovarian cancer cells B-57.In summary our investigations indicate that even in multi-passaged cancer cell lines hierarchic government of growth and differentiation is conserved and that the key cancer stem cell population may be composed of small overlapping cell fractions defined by various arbitrary markers.


Asunto(s)
Células Madre Neoplásicas/fisiología , Neoplasias Ováricas/patología , Animales , Separación Celular , Técnicas de Cocultivo , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Endonucleasas/genética , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico
5.
Oncogene ; 36(44): 6190-6203, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28869600

RESUMEN

Forkhead box O class transcription factors are homeostasis regulators that control cell death, longevity and therapy-resistance. In neuroblastoma (NB), nuclear FOXO3 correlates with stage M disease and poor prognosis. To analyze whether FOXO3 contributes to drug-resistance in this childhood cancer, we investigated how different high-stage-derived NB cells respond to the activation of an ectopic FOXO3 allele. We found endogenous FOXO3 mostly localized to the nucleus-upon activation of an ectopic, 4OHT-activated FOXO3(A3)ER fusion protein two of the cell lines underwent apoptosis, whereas in the others FOXO3-activation even increased survival during drug-treatment. In the latter cell type, FOXO3 did not induce the BH3-only protein BCL2L11/BIM due to impaired binding of FOXO3 to the BIM-promoter, but still activated other FOXO3 targets. It was shown before that FOXO3 and TP53 physically interact with each other at two different regions-the TP53-N-terminus binds to the FOXO3-DNA binding domain (DBD) and the FOXO3-C-terminus interacts with the TP53-DBD. Interestingly, cell lines that undergo FOXO3-induced cell death carry homozygous point mutations in the TP53-DBD near the structural hotspot-mutation-site R175H, which abrogated FOXO3-TP53 interaction. In contrast, in FOXO3-death-resistant cells no point mutations in the TP53-DBD were found-in these cells FOXO3-TP53 complexes are formed and FOXO3-binding to the BIM-promoter, but not the induction of the detoxifying protein SESN3, were prevented, which in turn increased chemo-protection in this type of high-stage-derived NB cells. Our combined data suggest that FOXO3 steps in as a death inducer in case of TP53-mutation, whereas functional TP53 alters FOXO3-target-promoter-recognition, which prevents death induction by FOXO3 and instead increases chemo-protection and survival of NB cells. This novel mechanism may explain the low incidence of TP53 mutation in high-stage NB at diagnosis and suggests FOXO3 as a therapeutic target for this childhood malignancy.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Proteína Forkhead Box O3/genética , Proteínas de Choque Térmico/genética , Neuroblastoma/genética , Proteína p53 Supresora de Tumor/genética , Apoptosis/genética , Línea Celular Tumoral , Núcleo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/genética , Proteína Forkhead Box O3/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Mutación , Estadificación de Neoplasias , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Regiones Promotoras Genéticas , Unión Proteica , Proteína p53 Supresora de Tumor/metabolismo
6.
Methods Inf Med ; 44(4): 516-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16342918

RESUMEN

OBJECTIVES: Changes in the status of DNA methylation, known as epigenetic alterations, are among the most common molecular alterations in human neoplasia. For the first time, we reported on the analysis of fecal DNA from patients with CRC to determine the feasibility, sensitivity and specificity of this approach. We want to present basic information about DNA methylation analysis in the context of bioinformatics, the study design and several statistical experiences with gene methylation data. Additionally we outline chances and new research questions in the field of DNA methylation. METHODS: We present current approaches to DNA methylation analysis based on one reference study. Its study design and the statistical analysis is reflected in the context of biomarker development. Finally we outline perspectives and research questions for statisticians and bioinformaticians. RESULTS: Identification of at least three genes as potential DNA methylation-based tumor marker genes (SFRP2, SFRP5, PGR). CONCLUSIONS: DNA methylation analysis is a rising topic in molecular genetics. Gene methylation will push the extension of biobanks to include new types of genetic data. Study design and statistical methods for the detection of methylation biomarkers must be improved. For the purpose of establishing methylation analysis as a new diagnostic/prognostic tool the adaptation of several approaches has become a challenging field of research activity.


Asunto(s)
Neoplasias del Colon/genética , Biología Computacional , Metilación de ADN , Epigénesis Genética , Biomarcadores de Tumor , Estudios de Factibilidad , Humanos , Sensibilidad y Especificidad
7.
Br J Pharmacol ; 172(11): 2748-55, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25073649

RESUMEN

The perioperative period is characterized by profound changes in the body's homoeostatic processes. This review seeks to address whether epigenetic mechanisms may influence an individual's reaction to surgery and anaesthesia. Evidence from animal and human studies suggests that epigenetic mechanisms can explain many facets of susceptibility to acute and chronic pain, making them potential therapeutic targets. Modern pain management is still based upon opiates, and both the developmental expression of opioid receptors and opioid-induced hyperalgesia have been linked to epigenetic mechanisms. In general, opiates seem to increase global DNA methylation levels. This is in contrast to local anaesthetics, which have been ascribed a global demethylating effect. Even though no direct investigations have been carried out, the potential influence of epigenetics on the inflammatory response that follows surgery seems a promising area for research. There is a considerable body of evidence that supports the involvement of epigenetics in the complex process of wound healing. Epigenetics is an important emerging research topic in perioperative medicine, with a huge potential to positively influence patient outcome.


Asunto(s)
Periodo de Recuperación de la Anestesia , Epigénesis Genética/genética , Inflamación/genética , Dolor Postoperatorio/genética , Periodo Perioperatorio , Estrés Fisiológico/genética , Cicatrización de Heridas/genética , Analgésicos Opioides/farmacología , Anestésicos Locales/farmacología , Metilación de ADN , Epigénesis Genética/efectos de los fármacos , Humanos , Hiperalgesia/genética , Dolor Postoperatorio/tratamiento farmacológico
8.
Cell Death Differ ; 19(8): 1277-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22322859

RESUMEN

Transforming growth factor beta (TGF-ß)- and Interleukin-2 (IL-2)-mediated signaling enables the generation and expansion of induced regulatory T (iTreg) cells that carry high hopes for the treatment of chronic inflammatory and autoimmune diseases. Knowledge about factors stabilizing their lineage commitment and lifespan, however, is limited. Here, we investigated the behavior of iTreg cells, derived from apoptosis-defective mouse mutants, during activated cell autonomous cell death, triggered by cytokine-deprivation, or activation-induced cell death (AICD) after restimulation of the T-cell receptor, and compared these responses with those of effector T cells. We observed that iTreg cells were much more sensitive to IL-2-deprivation but poorly susceptible to AICD. In fact, when apoptosis was compromised, T-cell receptor (TCR)-religation resulted in methylation-independent, ERK- and PI3K/mTOR-mediated loss of Foxp3 expression, impaired suppressive capacity and effector cytokine production. Although iTreg cells prevented colitis induction they rapidly lost Foxp3-GFP expression and gained ability to produce effector cytokines thereby imposing Th1 cell fate on resident effector cells. Surprisingly, iTreg cell conversion itself was limited by TGF-ß-mediated Bim/Bcl2L11-dependent apoptosis. Hence, the very same cytokine that drives the generation of iTreg cells can trigger their demise. Our results provide novel insights in iTreg cell biology that will assist optimization of iTreg-based therapy.


Asunto(s)
Interleucina-2/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Apoptosis/inmunología , Supervivencia Celular/inmunología , Colitis/inmunología , Proteína Ligando Fas/inmunología , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/inmunología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptor fas/deficiencia , Receptor fas/inmunología
9.
Oncogene ; 30(38): 4038-49, 2011 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-21516127

RESUMEN

Recently we showed an integral epidermal growth factor receptor (EGFR)-E2F3a signaling path, in which E2F3a was found to be essential in EGFR-mediated proliferation in ovarian cancer cells. The present work evaluates the clinical relevance of this novel axis and of E2F3a itself in a large set of 130 ovarian cancer specimens. For this purpose E2F3a and its counterpart, E2F3b, were measured by RT-PCR and activated EGFR was assessed by immunohistochemistry. When compared with healthy control tissue, both E2F3 isoforms were overexpressed in the cancers, but only E2F3a expression correlated with tumor stage (ρ=0.349, P=0.0001) and residual disease (ρ=0.254, P=0.004). Univariate survival analyses showed E2F3a and activated EGFR to be associated with poor PFS and OS. Furthermore, a strong, positive correlation between activated EGFR and E2F3a expression was shown (P=0.0001). We further identified two EGFR-independent mechanisms that regulate E2F3a expression, namely one, acting by promoter methylation of miR-34a, which by its physical interaction with E2F3a transcripts causes their degradation, and the second based on 6p22 gene locus amplification. MiRIDIAN-based knockdown and induction of miR-34a evidenced a direct regulatory link between miR-34a and E2F3a, and the tumor-suppressive character of miR-34a was documented by its association with improved survival. Although, 6p22 gene locus amplification was detected in a significant number of ovarian cancer specimens, 6p22 ploidy was not relevant in predicting survival. In Cox regression analysis, E2F3a, but not activated EGFR or miR-34a expression, retained independent prognostic significance (PFS: hazards ratio 3.785 (1.326-9.840), P=0.013; OS: hazards ratio 4.651 (1.189-15.572), P=0.013). These clinical findings highlight the relevance of E2F3a in the biology of ovarian cancer. Moreover, identification of EGFR-independent mechanisms in E2F3a control can be helpful in explaining the non-responsiveness of therapeutic EGFR targeting in ovarian cancer.


Asunto(s)
Factor de Transcripción E2F3/fisiología , Neoplasias Ováricas/patología , Anciano , Cromosomas Humanos Par 6 , Metilación de ADN , Factor de Transcripción E2F3/análisis , Factor de Transcripción E2F3/genética , Receptores ErbB/fisiología , Femenino , Amplificación de Genes , Humanos , MicroARNs/genética , Persona de Mediana Edad , Neoplasias Ováricas/química , Neoplasias Ováricas/genética , Pronóstico , Regiones Promotoras Genéticas
10.
Oncogene ; 29(13): 1997-2004, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20101229

RESUMEN

Similar to p73, the tumor suppressor gene p53 is subject to alternative splicing. Besides p53DeltaE6 and p53beta, we identified p53zeta, p53delta and p53varepsilon, arising from alternative splicing of exon 6 and intron 9, respectively. p53 splice variants were present in 18 of 34 ovarian cancer cell lines (52.9%) and 134 of 245 primary ovarian cancers (54.7%). p53delta expression was associated with impaired response to primary platinum-based chemotherapy (P=0.032). Also, p53delta expression constituted an independent prognostic marker for recurrence-free and overall survival (hazard ratio 1.854, 95% confidence interval 1.121-3.065, P=0.016; and hazard ratio 1.937, 95% confidence interval 1.177-3.186, P=0.009, respectively). p53beta expression was associated with adverse clinicopathologic markers, that is, serous and poorly differentiated cancers (P=0.002 and P=0.008, respectively), and correlated with worse recurrence-free survival in patients exhibiting functionally active p53 (P=0.049). DeltaN'p73 constituted the main N-terminally truncated p73 isoform and was preferentially found in ovarian cancer cell lines showing functionally active p53, supporting our hypothesis that N-terminally truncated p73 isoforms can alleviate the selection pressure for p53 mutations by the inhibition of p53 protein function.


Asunto(s)
Empalme Alternativo , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Femenino , Humanos , Intrones , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/metabolismo
11.
Br J Cancer ; 89(10): 1934-9, 2003 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-14612906

RESUMEN

This study analysed mRNA expression of two members of the methyl-CpG-binding protein family - MeCP2 and MBD2 - in human non-neoplastic (n=11) and neoplastic (n=57) breast tissue specimens using a quantitative real-time PCR method. We observed higher expression levels of MeCP2 mRNA in neoplastic tissues than in non-neoplastic tissues (P=0.001), whereas no significant differences for MBD2 were detected. When studying the relations between the most important clinicopathologic features of breast cancer and the mRNA expression level of both MBDs, we found that oestrogen receptor (OR)-positive breast cancer specimens contained higher levels of MeCP2 mRNA than did OR-negative cancers (P=0.005). Furthermore, we observed statistically significantly higher levels of MeCP2 in non-neoplastic tissues expressing high levels of OR as compared to those expressing low levels (P=0.017). Finally, using a linear regression model, we identified a statistically significant association between OR expression and MeCP2 mRNA expression in neoplastic and non-neoplastic breast tissue specimens (P=0.003). In conclusion, we were able to demonstrate for the first time that there exists a strong association between OR status and MeCP2 mRNA expression. Furthermore, we speculate that MeCP2, regulated by OR, plays a key role in the differentiation processes in human breast tissues.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/biosíntesis , Regulación Neoplásica de la Expresión Génica , Receptores de Estrógenos/análisis , Adulto , Mama/fisiología , Diferenciación Celular , Islas de CpG , Proteínas de Unión al ADN/farmacología , Femenino , Humanos , Proteína 2 de Unión a Metil-CpG , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Análisis de Regresión , Proteínas Represoras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA