Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Ther Nucleic Acids ; 32: 773-793, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37346977

RESUMEN

Antisense oligonucleotide (ASO) therapeutics are being investigated for a broad range of neurological diseases. While ASOs have been effective in the clinic, improving productive ASO internalization into target cells remains a key area of focus in the field. Here, we investigated how the delivery of ASO-loaded lipid nanoparticles (LNPs) affects ASO activity, subcellular trafficking, and distribution in the brain. We show that ASO-LNPs increase ASO activity up to 100-fold in cultured primary brain cells as compared to non-encapsulated ASO. However, in contrast to the widespread ASO uptake and activity observed following free ASO delivery in vivo, LNP-delivered ASOs did not downregulate mRNA levels throughout the brain after intracerebroventricular injection. This lack of activity was likely due to ASO accumulation in cells lining the ventricles and blood vessels. Furthermore, we reveal a formulation-dependent activation of the immune system post dosing, suggesting that LNP encapsulation cannot mask cellular ASO backbone-mediated toxicities. Together, these data provide insights into how LNP encapsulation affects ASO distribution as well as activity in the brain, and a foundation that enables future optimization of brain-targeting ASO-LNPs.

2.
J Vis Exp ; (185)2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35969105

RESUMEN

Positive and negative controls with known expression of target proteins are essential for the development of immunohistochemistry (IHC) assays. While tissue controls are beneficial for well-characterized proteins with defined tissue and cellular expression patterns, they are less suitable for the initial development of IHC assays for novel, poorly characterized, or ubiquitously expressed proteins. Alternatively, due to their standardized nature, cell pellets, including cancer cell lines with defined protein or transcript expression levels (e.g., high, medium, and low expression), transfected over-expressing cell lines, or cell lines with genes deleted through cell engineering technologies like CRISPR, can serve as valuable controls, especially for the initial antibody characterization and selection. In order for these cell pellets to be used in the development of IHC assays for formalin-fixed, paraffin-embedded tissues, they need to be processed and embedded in a manner that recapitulates the procedures used for tissue processing. This protocol describes a process for creating and processing formalin-fixed, paraffin-embedded cell pellet controls that can be used for IHC method developments.


Asunto(s)
Formaldehído , Proteínas , Formaldehído/química , Inmunohistoquímica , Adhesión en Parafina , Fijación del Tejido/métodos
3.
J Vis Exp ; (174)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34487120

RESUMEN

Immunohistochemistry (IHC) assays provide valuable insights into protein expression patterns, the reliable interpretation of which requires well-characterized positive and negative control samples. Because appropriate tissue or cell line controls are not always available, a simple method to create synthetic IHC controls may be beneficial. Such a method is described here. It is adaptable to various antigen types, including proteins, peptides, or oligonucleotides, in a wide range of concentrations. This protocol explains the steps necessary to create synthetic antigen controls, using as an example a peptide from the human erythroblastic oncogene B2 (ERBB2/HER2) intracellular domain (ICD) recognized by a variety of diagnostically relevant antibodies. Serial dilutions of the HER2 ICD peptide in bovine serum albumin (BSA) solution are mixed with formaldehyde and heated for 10 min at 85 °C to solidify and cross-link the peptide/BSA mixture. The resulting gel can be processed, sectioned, and stained like a tissue, yielding a series of samples of known antigen concentrations spanning a wide range of staining intensities. This simple protocol is consistent with routine histology lab procedures. The method requires only that the user have a sufficient quantity of the desired antigen. Recombinant proteins, protein domains, or linear peptides that encode relevant epitopes may be synthesized locally or commercially. Laboratories generating in-house antibodies can reserve aliquots of the immunizing antigen as the synthetic control target. The opportunity to create well-defined positive controls across a wide range of concentrations allows users to assess intra- and inter-laboratory assay performance, gain insight into the dynamic range and linearity of their assays, and optimize assay conditions for their particular experimental goals.


Asunto(s)
Antígenos , Formaldehído , Epítopos , Humanos , Inmunohistoquímica , Vacunas Sintéticas
4.
Mol Cancer Ther ; 20(4): 716-725, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33536191

RESUMEN

Ovarian cancer is a diverse class of tumors with very few effective treatment options and suboptimal response rates in early clinical studies using immunotherapies. Here we describe LY6/PLAUR domain containing 1 (LYPD1) as a novel target for therapeutic antibodies for the treatment of ovarian cancer. LYPD1 is broadly expressed in both primary and metastatic ovarian cancer with ∼70% prevalence in the serous cancer subset. Bispecific antibodies targeting CD3 on T cells and a tumor antigen on cancer cells have demonstrated significant clinical activity in hematologic cancers. We have developed an anti-LYPD1/CD3 T-cell-dependent bispecific antibody (TDB) to redirect T-cell responses to LYPD1 expressing ovarian cancer. Here we characterize the nonclinical pharmacology of anti-LYPD1/CD3 TDB and show induction of a robust polyclonal T-cell activation and target dependent killing of LYPD1 expressing ovarian cancer cells resulting in efficient in vivo antitumor responses in PBMC reconstituted immune-deficient mice and human CD3 transgenic mouse models. Anti-LYPD1/CD3 TDB is generally well tolerated at high-dose levels in mice, a pharmacologically relevant species, and showed no evidence of toxicity or damage to LYPD1 expressing tissues.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3/inmunología , Neoplasias Ováricas/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Anticuerpos Biespecíficos/farmacología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Transgénicos , Neoplasias Ováricas/patología
5.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33620419

RESUMEN

Despite the development of effective therapies, a substantial proportion of asthmatics continue to have uncontrolled symptoms, airflow limitation, and exacerbations. Transient receptor potential cation channel member A1 (TRPA1) agonists are elevated in human asthmatic airways, and in rodents, TRPA1 is involved in the induction of airway inflammation and hyperreactivity. Here, the discovery and early clinical development of GDC-0334, a highly potent, selective, and orally bioavailable TRPA1 antagonist, is described. GDC-0334 inhibited TRPA1 function on airway smooth muscle and sensory neurons, decreasing edema, dermal blood flow (DBF), cough, and allergic airway inflammation in several preclinical species. In a healthy volunteer Phase 1 study, treatment with GDC-0334 reduced TRPA1 agonist-induced DBF, pain, and itch, demonstrating GDC-0334 target engagement in humans. These data provide therapeutic rationale for evaluating TRPA1 inhibition as a clinical therapy for asthma.


Asunto(s)
Asma/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Dolor/tratamiento farmacológico , Prurito/tratamiento farmacológico , Piridinas/farmacología , Piridinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Canal Catiónico TRPA1/antagonistas & inhibidores , Adolescente , Adulto , Animales , Estudios de Cohortes , Modelos Animales de Enfermedad , Perros , Método Doble Ciego , Femenino , Cobayas , Voluntarios Sanos , Humanos , Isotiocianatos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Dolor/inducido químicamente , Prurito/inducido químicamente , Ratas , Ratas Sprague-Dawley , Canal Catiónico TRPA1/deficiencia , Resultado del Tratamiento , Adulto Joven
6.
J Am Soc Nephrol ; 31(9): 2044-2064, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32764142

RESUMEN

BACKGROUND: APOL1 is found in human kidney podocytes and endothelia. Variants G1 and G2 of the APOL1 gene account for the high frequency of nondiabetic CKD among African Americans. Proposed mechanisms of kidney podocyte cytotoxicity resulting from APOL1 variant overexpression implicate different subcellular compartments. It is unclear where endogenous podocyte APOL1 resides, because previous immunolocalization studies utilized overexpressed protein or commercially available antibodies that crossreact with APOL2. This study describes and distinguishes the locations of both APOLs. METHODS: Immunohistochemistry, confocal and immunoelectron microscopy, and podocyte fractionation localized endogenous and transfected APOL1 using a large panel of novel APOL1-specific mouse and rabbit monoclonal antibodies. RESULTS: Both endogenous podocyte and transfected APOL1 isoforms vA and vB1 (and a little of isoform vC) localize to the luminal face of the endoplasmic reticulum (ER) and to the cell surface, but not to mitochondria, endosomes, or lipid droplets. In contrast, APOL2, isoform vB3, and most vC of APOL1 localize to the cytoplasmic face of the ER and are consequently absent from the cell surface. APOL1 knockout podocytes do not stain for APOL1, attesting to the APOL1-specificity of the antibodies. Stable re-transfection of knockout podocytes with inducible APOL1-G0, -G1, and -G2 showed no differences in localization among variants. CONCLUSIONS: APOL1 is found in the ER and plasma membrane, consistent with either the ER stress or surface cation channel models of APOL1-mediated cytotoxicity. The surface localization of APOL1 variants potentially opens new therapeutic targeting avenues.


Asunto(s)
Apolipoproteína L1/análisis , Membrana Celular/química , Retículo Endoplásmico/química , Podocitos/química , Animales , Anticuerpos/inmunología , Apolipoproteína L1/inmunología , Apolipoproteínas L/análisis , Células COS , Células Cultivadas , Chlorocebus aethiops , Reacciones Cruzadas , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Podocitos/ultraestructura
7.
J Histochem Cytochem ; 67(5): 309-334, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30879407

RESUMEN

Optimization and standardization of immunohistochemistry (IHC) protocols within and between laboratories requires reproducible positive and negative control samples. In many situations, suitable tissue or cell line controls are not available. We demonstrate here a method to incorporate target antigens into synthetic protein gels that can serve as IHC controls. The method can use peptides, protein domains, or whole proteins as antigens, and is compatible with a variety of fixation protocols. The resulting gels can be used to create tissue microarrays (TMAs) with a range of antigen concentrations that can be used to objectively quantify and calibrate chromogenic, fluorescent, or mass spectrometry-based IHC protocols. The method offers an opportunity to objectively quantify IHC staining results, and to optimize and standardize IHC protocols within and between laboratories. (J Histochem Cytochem 58:XXX-XXX, 2019).


Asunto(s)
Antígenos/análisis , Geles/química , Inmunohistoquímica/métodos , Animales , Formaldehído/química , Humanos , Inmunohistoquímica/normas , Ratones , Coloración y Etiquetado/métodos , Coloración y Etiquetado/normas , Análisis de Matrices Tisulares/métodos , Análisis de Matrices Tisulares/normas , Fijación del Tejido/métodos , Fijación del Tejido/normas
8.
Pigment Cell Melanoma Res ; 32(2): 269-279, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30156010

RESUMEN

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk of mesothelioma and melanocytic tumors. Here, we show that Bap1 deletion in melanocytes cooperates with the constitutively active, oncogenic form of BRAF (BRAFV600E ) and UV to cause melanoma in mice, albeit at very low frequency. In addition, Bap1-null melanoma cells derived from mouse tumors are more aggressive and colonize and grow at distant sites more than their wild-type counterparts. Molecularly, Bap1-null melanoma cell lines have increased DNA damage measured by γH2aX and hyperubiquitination of histone H2a. Therapeutically, these Bap1-null tumors are completely responsive to BRAF- and MEK-targeted therapies. Therefore, BAP1 functions as a tumor suppressor and limits tumor progression in melanoma.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Melanoma/genética , Melanoma/patología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , Transición Epitelial-Mesenquimal/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Melanocitos/metabolismo , Melanocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Transcripción Genética , Ubiquitinación , Melanoma Cutáneo Maligno
9.
Hepatology ; 61(3): 942-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25311838

RESUMEN

UNLABELLED: Primary liver cancer encompasses both hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). The Notch signaling pathway, known to be important for the proper development of liver architecture, is also a potential driver of primary liver cancer. However, with four known Notch receptors and several Notch ligands, it is not clear which Notch pathway members play the predominant role in liver cancer. To address this question, we utilized antibodies to specifically target Notch1, Notch2, Notch3, or jagged1 (Jag1) in a mouse model of primary liver cancer driven by v-akt murine thymoma viral oncogene homolog and neuroblastoma RAS viral oncogene homolog (NRas). We show that inhibition of Notch2 reduces tumor burden by eliminating highly malignant HCC- and CCA-like tumors. Inhibition of the Notch ligand, Jag1, had a similar effect, consistent with Jag1 acting in cooperation with Notch2. This effect was specific to Notch2, because Notch3 inhibition did not decrease tumor burden. Unexpectedly, Notch1 inhibition altered the relative proportion of tumor types, reducing HCC-like tumors but dramatically increasing CC-like tumors. Finally, we show that Notch2 and Jag1 are expressed in, and Notch2 signaling is activated in, a subset of human HCC samples. CONCLUSIONS: These findings underscore the distinct roles of different Notch receptors in the liver and suggest that inhibition of Notch2 signaling represents a novel therapeutic option in the treatment of liver cancer.


Asunto(s)
Neoplasias Hepáticas/tratamiento farmacológico , Receptores Notch/antagonistas & inhibidores , Animales , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/antagonistas & inhibidores , Modelos Animales de Enfermedad , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/análisis , Humanos , Péptidos y Proteínas de Señalización Intercelular/análisis , Proteína Jagged-1 , Proteínas de la Membrana/análisis , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Receptores Notch/análisis , Receptores Notch/fisiología , Proteínas Serrate-Jagged
10.
PLoS One ; 7(5): e36713, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615798

RESUMEN

The fibroblast growth factor (FGF)-FGF receptor (FGFR) signaling system plays critical roles in a variety of normal developmental and physiological processes. It is also well documented that dysregulation of FGF-FGFR signaling may have important roles in tumor development and progression. The FGFR4-FGF19 signaling axis has been implicated in the development of hepatocellular carcinomas (HCCs) in mice, and potentially in humans. In this study, we demonstrate that FGFR4 is required for hepatocarcinogenesis; the progeny of FGF19 transgenic mice, which have previously been shown to develop HCCs, bred with FGFR4 knockout mice fail to develop liver tumors. To further test the importance of FGFR4 in HCC, we developed a blocking anti-FGFR4 monoclonal antibody (LD1). LD1 inhibited: 1) FGF1 and FGF19 binding to FGFR4, 2) FGFR4-mediated signaling, colony formation, and proliferation in vitro, and 3) tumor growth in a preclinical model of liver cancer in vivo. Finally, we show that FGFR4 expression is elevated in several types of cancer, including liver cancer, as compared to normal tissues. These findings suggest a modulatory role for FGFR4 in the development and progression of hepatocellular carcinoma and that FGFR4 may be an important and novel therapeutic target in treating this disease.


Asunto(s)
Carcinoma Hepatocelular/prevención & control , Modelos Animales de Enfermedad , Neoplasias Hepáticas/prevención & control , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Animales , Anticuerpos Neutralizantes/inmunología , Carcinoma Hepatocelular/patología , División Celular , Neoplasias Hepáticas/patología , Ratones , Ratones Transgénicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/inmunología
11.
Toxicol Sci ; 126(2): 446-56, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22268002

RESUMEN

Fibroblast growth factor 19 (FGF19) represses cholesterol 7α-hydroxylase (Cyp7α1) and inhibits bile acid synthesis in vitro and in vivo. Previous studies have shown that anti-FGF19 antibody treatment reduces growth of colon tumor xenografts and prevents hepatocellular carcinomas in FGF19 transgenic mice and thus may be a useful cancer target. In a repeat dose safety study in cynomolgus monkeys, anti-FGF19 treatment (3-100 mg/kg) demonstrated dose-related liver toxicity accompanied by severe diarrhea and low food consumption. The mechanism of anti-FGF19 toxicity was investigated using in vitro and in vivo approaches. Our results show that anti-FGF19 antibody had no direct cytotoxic effect on monkey hepatocytes. Anti-FGF19 increased Cyp7α1, as expected, but also increased bile acid efflux transporter gene (bile salt export pump, multidrug resistant protein 2 [MRP2], and MRP3) expression and reduced sodium taurocholate cotransporting polypeptide and organic anion transporter 2 expression in liver tissues from treated monkeys and in primary hepatocytes. In addition, anti-FGF19 treatment increased solute transporter gene (ileal bile acid-binding protein, organic solute transporter α [OST-α], and OST-ß) expression in ileal tissues from treated monkeys but not in Caco-2 cells. However, deoxycholic acid (a secondary bile acid) increased expression of FGF19 and these solute transporter genes in Caco-2 cells. Gas chromatography-mass spectrometry analysis of monkey feces showed an increase in total bile acids and cholic acid derivatives. These findings suggest that high doses of anti-FGF19 increase Cyp7α1 expression and bile acid synthesis and alter the expression of bile transporters in the liver resulting in enhanced bile acid efflux and reduced uptake. Increased bile acids alter expression of solute transporters in the ileum causing diarrhea and the enhanced enterohepatic recirculation of bile acids leading to liver toxicity.


Asunto(s)
Anticuerpos/inmunología , Ácidos y Sales Biliares/biosíntesis , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Íleon/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Factores de Crecimiento de Fibroblastos/inmunología , Perfilación de la Expresión Génica , Hepatocitos/citología , Hepatocitos/metabolismo , Absorción Intestinal , Macaca fascicularis
12.
Cancer Res ; 68(13): 5086-95, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18593907

RESUMEN

Fibroblast growth factors (FGF) play important roles in development, angiogenesis, and cancer. FGF19 uniquely binds to FGF receptor 4 (FGFR4). Our previous study has shown that FGF19 transgenic tumors have an activated Wnt-pathway phenotype. Wnt signaling is implicated in initiating or promoting FGF signaling in various cell types and organs. In this study, we examined whether FGF19 or inhibition of FGF19 affects the beta-catenin signaling pathway using human colon cancer cell lines (HCT116, Colo201). Our results show that FGF19 increases tyrosine phosphorylation of beta-catenin and causes loss of beta-catenin-E-cadherin binding. FGF19 increases p-GSK3beta and active beta-catenin levels and anti-FGF19 antibody (1A6) treatment abrogates this effect of FGF19. Anti-FGF19 antibody treatment increases S33/S37/T41 phosphorylation and ubiquitination of beta-catenin. Ion-trap mass spectrometric analysis confirmed that 1A6 increases phosphorylation of beta-catenin in the NH(2) terminus. Using HCT116-paired beta-catenin knockout cells, we show that FGF19 induces TCF/LEF reporter activity in parental (WT/Delta45) and in WT/--but not in mutant (-/Delta45) cells, and that inhibition of endogenous FGF19 reduces this reporter activity, indicating that wild-type beta-catenin is accessible for modulation. FGFR4 knockdown using inducible short hairpin RNA significantly reduces the colony-forming ability in vitro and tumor growth in vivo. Although cleaved caspase-3 immunoreactivity remains unchanged, the number of ki67-positive nuclei is reduced in FGFR4 knockdown tumor xenograft tissues. Consistent with the reduced beta-catenin activation, Taqman analyses show that FGF19/FGFR4 inhibition reduced beta-catenin target gene (cyclin D1, CD44, c-jun, Cox-2, UPAR) expression. These findings highlight that FGF19/FGFR4 cross-talk with beta-catenin and that pathway intervention reduces tumor growth.


Asunto(s)
Anticuerpos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , beta Catenina/metabolismo , Animales , Dominio Catalítico , Femenino , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/inmunología , Factores de Crecimiento de Fibroblastos/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , ARN Interferente Pequeño/farmacología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Treonina/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Virology ; 325(1): 129-36, 2004 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-15231392

RESUMEN

This study evaluated the hypothesis that the disease status of Saanen goats infected with caprine arthritis-encephalitis lentivirus (CAEV) is associated with the focus of immune responses to viral antigens, particularly the surface envelope glycoprotein (SU). Specifically, we have proposed that Th2 responses promote progressive immune-mediated arthritis, whereas Th1 responses restrict virus replication and development of clinical disease. The present study determined the isotype of SU antibodies associated with progressor and long-term nonprogressor (LTNP) status. We show that chronically infected goats that develop clinical arthritis have predominantly IgG1 antibodies to SU during both preclinical and clinical stages of disease, whereas SU antibodies of LTNP goats are relatively biased toward IgG2. Additional studies determined the isotype of SU antibodies induced initially by CAEV infection. These experiments show that initial IgG1-dominated responses to SU are associated with subsequent development of preclinical inflammatory joint lesions, whereas lack of joint pathology is associated with an IgG2 bias of initial responses to SU. Our results using the CAEV model suggest that isotype bias of SU antibodies is a reliable indicator of clinical disease caused by lentiviruses. Isotype analysis may be a useful method to screen candidate lentiviral vaccines intended to prevent disease progression.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus de la Artritis-Encefalitis Caprina/inmunología , Productos del Gen env/inmunología , Isotipos de Inmunoglobulinas/sangre , Infecciones por Lentivirus/inmunología , Animales , Cabras , Inmunoglobulina G/sangre , Inmunoglobulina G/clasificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA