Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 622(7982): 329-338, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37794186

RESUMEN

The Pharma Proteomics Project is a precompetitive biopharmaceutical consortium characterizing the plasma proteomic profiles of 54,219 UK Biobank participants. Here we provide a detailed summary of this initiative, including technical and biological validations, insights into proteomic disease signatures, and prediction modelling for various demographic and health indicators. We present comprehensive protein quantitative trait locus (pQTL) mapping of 2,923 proteins that identifies 14,287 primary genetic associations, of which 81% are previously undescribed, alongside ancestry-specific pQTL mapping in non-European individuals. The study provides an updated characterization of the genetic architecture of the plasma proteome, contextualized with projected pQTL discovery rates as sample sizes and proteomic assay coverages increase over time. We offer extensive insights into trans pQTLs across multiple biological domains, highlight genetic influences on ligand-receptor interactions and pathway perturbations across a diverse collection of cytokines and complement networks, and illustrate long-range epistatic effects of ABO blood group and FUT2 secretor status on proteins with gastrointestinal tissue-enriched expression. We demonstrate the utility of these data for drug discovery by extending the genetic proxied effects of protein targets, such as PCSK9, on additional endpoints, and disentangle specific genes and proteins perturbed at loci associated with COVID-19 susceptibility. This public-private partnership provides the scientific community with an open-access proteomics resource of considerable breadth and depth to help to elucidate the biological mechanisms underlying proteo-genomic discoveries and accelerate the development of biomarkers, predictive models and therapeutics1.


Asunto(s)
Bancos de Muestras Biológicas , Proteínas Sanguíneas , Bases de Datos Factuales , Genómica , Salud , Proteoma , Proteómica , Humanos , Sistema del Grupo Sanguíneo ABO/genética , Proteínas Sanguíneas/análisis , Proteínas Sanguíneas/genética , COVID-19/genética , Descubrimiento de Drogas , Epistasis Genética , Fucosiltransferasas/metabolismo , Predisposición Genética a la Enfermedad , Plasma/química , Proproteína Convertasa 9/metabolismo , Proteoma/análisis , Proteoma/genética , Asociación entre el Sector Público-Privado , Sitios de Carácter Cuantitativo , Reino Unido , Galactósido 2-alfa-L-Fucosiltransferasa
2.
Neuropsychopharmacology ; 47(7): 1387-1397, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34593976

RESUMEN

Tianeptine is an atypical antidepressant used in Europe to treat patients who respond poorly to selective serotonin reuptake inhibitors (SSRIs). The recent discovery that tianeptine is a mu opioid receptor (MOR) agonist has provided a potential avenue for expanding our understanding of antidepressant treatment beyond the monoamine hypothesis. Thus, our studies aim to understand the neural circuits underlying tianeptine's antidepressant effects. We show that tianeptine induces rapid antidepressant-like effects in mice after as little as one week of treatment. Critically, we also demonstrate that tianeptine's mechanism of action is distinct from fluoxetine in two important aspects: (1) tianeptine requires MORs for its chronic antidepressant-like effect, while fluoxetine does not, and (2) unlike fluoxetine, tianeptine does not promote hippocampal neurogenesis. Using cell-type specific MOR knockouts we further show that MOR expression on GABAergic cells-specifically somatostatin-positive neurons-is necessary for the acute and chronic antidepressant-like responses to tianeptine. Using central infusion of tianeptine, we also implicate the ventral hippocampus as a potential site of antidepressant action. Moreover, we show a dissociation between the antidepressant-like phenotype and other opioid-like phenotypes resulting from acute tianeptine administration such as analgesia, conditioned place preference, and hyperlocomotion. Taken together, these results suggest a novel entry point for understanding what circuit dysregulations may occur in depression, as well as possible targets for the development of new classes of antidepressant drugs.


Asunto(s)
Receptores Opioides mu , Tiazepinas , Analgésicos Opioides/farmacología , Animales , Antidepresivos/farmacología , Fluoxetina/farmacología , Hipocampo , Humanos , Interneuronas , Ratones , Receptores Opioides mu/agonistas , Tiazepinas/farmacología
3.
Elife ; 102021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33555255

RESUMEN

Controlling receptor functional selectivity profiles for opioid receptors is a promising approach for discovering safer analgesics; however, the structural determinants conferring functional selectivity are not well understood. Here, we used crystal structures of opioid receptors, including the recently solved active state kappa opioid complex with MP1104, to rationally design novel mixed mu (MOR) and kappa (KOR) opioid receptor agonists with reduced arrestin signaling. Analysis of structure-activity relationships for new MP1104 analogs points to a region between transmembrane 5 (TM5) and extracellular loop (ECL2) as key for modulation of arrestin recruitment to both MOR and KOR. The lead compounds, MP1207 and MP1208, displayed MOR/KOR Gi-partial agonism with diminished arrestin signaling, showed efficient analgesia with attenuated liabilities, including respiratory depression and conditioned place preference and aversion in mice. The findings validate a novel structure-inspired paradigm for achieving beneficial in vivo profiles for analgesia through different mechanisms that include bias, partial agonism, and dual MOR/KOR agonism.


Asunto(s)
Morfinanos/química , Receptores Opioides kappa/química , Receptores Opioides mu/química , Secuencias de Aminoácidos , Analgésicos/química , Analgésicos/metabolismo , Animales , Sitios de Unión , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
4.
Cell Mol Neurobiol ; 41(5): 977-993, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32424771

RESUMEN

Mu opioid receptors (MOR-1) mediate the biological actions of clinically used opioids such as morphine, oxycodone, and fentanyl. The mu opioid receptor gene, OPRM1, undergoes extensive alternative splicing, generating multiple splice variants. One type of splice variants are truncated variants containing only six transmembrane domains (6TM) that mediate the analgesic action of novel opioid drugs such as 3'-iodobenzoylnaltrexamide (IBNtxA). Previously, we have shown that IBNtxA is a potent analgesic effective in a spectrum of pain models but lacks many side-effects associated with traditional opiates. In order to investigate the targets labeled by IBNtxA, we synthesized two arylazido analogs of IBNtxA that allow photolabeling of mouse mu opioid receptors (mMOR-1) in transfected cell lines and mMOR-1 protein complexes that may comprise the 6TM sites in mouse brain. We demonstrate that both allyl and alkyne arylazido derivatives of IBNtxA efficiently radio-photolabeled mMOR-1 in cell lines and MOR-1 protein complexes expressed either exogenously or endogenously, as well as found in mouse brain. In future, design and application of such radio-photolabeling ligands with a conjugated handle will provide useful tools for further isolating or purifying MOR-1 to investigate site specific ligand-protein contacts and its signaling complexes.


Asunto(s)
Analgésicos Opioides/metabolismo , Azidas/metabolismo , Encéfalo/metabolismo , Naltrexona/análogos & derivados , Etiquetas de Fotoafinidad/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/síntesis química , Animales , Azidas/síntesis química , Encéfalo/efectos de los fármacos , Células CHO , Línea Celular , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naltrexona/síntesis química , Naltrexona/metabolismo , Etiquetas de Fotoafinidad/síntesis química , Unión Proteica/fisiología , Ensayo de Unión Radioligante/métodos
5.
ACS Cent Sci ; 5(6): 992-1001, 2019 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-31263758

RESUMEN

Mitragyna speciosa, more commonly known as kratom, is a plant native to Southeast Asia, the leaves of which have been used traditionally as a stimulant, analgesic, and treatment for opioid addiction. Recently, growing use of the plant in the United States and concerns that kratom represents an uncontrolled drug with potential abuse liability, have highlighted the need for more careful study of its pharmacological activity. The major active alkaloid found in kratom, mitragynine, has been reported to have opioid agonist and analgesic activity in vitro and in animal models, consistent with the purported effects of kratom leaf in humans. However, preliminary research has provided some evidence that mitragynine and related compounds may act as atypical opioid agonists, inducing therapeutic effects such as analgesia, while limiting the negative side effects typical of classical opioids. Here we report evidence that an active metabolite plays an important role in mediating the analgesic effects of mitragynine. We find that mitragynine is converted in vitro in both mouse and human liver preparations to the much more potent mu-opioid receptor agonist 7-hydroxymitragynine and that this conversion is mediated by cytochrome P450 3A isoforms. Further, we show that 7-hydroxymitragynine is formed from mitragynine in mice and that brain concentrations of this metabolite are sufficient to explain most or all of the opioid-receptor-mediated analgesic activity of mitragynine. At the same time, mitragynine is found in the brains of mice at very high concentrations relative to its opioid receptor binding affinity, suggesting that it does not directly activate opioid receptors. The results presented here provide a metabolism-dependent mechanism for the analgesic effects of mitragynine and clarify the importance of route of administration for determining the activity of this compound. Further, they raise important questions about the interpretation of existing data on mitragynine and highlight critical areas for further research in animals and humans.

6.
Neuron ; 98(3): 575-587.e4, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29656874

RESUMEN

Dopamine input to the dorsal and ventral striatum originates from separate populations of midbrain neurons. Despite differences in afferent inputs and behavioral output, little is known about how dopamine release is encoded by dopamine receptors on medium spiny neurons (MSNs) across striatal subregions. Here we examined the activation of D2 receptors following the synaptic release of dopamine in the dorsal striatum (DStr) and nucleus accumbens (NAc) shell. We found that D2 receptor-mediated synaptic currents were slower in the NAc and this difference occurred at the level of D2-receptor signaling. As a result of preferential coupling to Gαo, we also found that D2 receptors in MSNs demonstrated higher sensitivity for dopamine in the NAc. The higher sensitivity in the NAc was eliminated following cocaine exposure. These results identify differences in the sensitivity and timing of D2-receptor signaling across the striatum that influence how nigrostriatal and mesolimbic signals are encoded across these circuits.


Asunto(s)
Cuerpo Estriado/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D2/metabolismo , Transducción de Señal/fisiología , Animales , Cuerpo Estriado/efectos de los fármacos , Dopamina/metabolismo , Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Núcleo Accumbens/efectos de los fármacos , Receptores de Dopamina D2/agonistas , Transducción de Señal/efectos de los fármacos
7.
Neuropsychopharmacology ; 42(10): 2052-2063, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28303899

RESUMEN

Depression is a debilitating chronic illness that affects around 350 million people worldwide. Current treatments, such as selective serotonin reuptake inhibitors, are not ideal because only a fraction of patients achieve remission. Tianeptine is an effective antidepressant with a previously unknown mechanism of action. We recently reported that tianeptine is a full agonist at the mu opioid receptor (MOR). Here we demonstrate that the acute and chronic antidepressant-like behavioral effects of tianeptine in mice require MOR. Interestingly, while tianeptine also produces many opiate-like behavioral effects such as analgesia and reward, it does not lead to tolerance or withdrawal. Furthermore, the primary metabolite of tianeptine (MC5), which has a longer half-life, mimics the behavioral effects of tianeptine in a MOR-dependent fashion. These results point to the possibility that MOR and its downstream signaling cascades may be novel targets for antidepressant drug development.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Receptores Opioides mu/metabolismo , Tiazepinas/farmacología , Analgésicos Opioides/farmacología , Animales , Antidepresivos Tricíclicos/metabolismo , Antidepresivos Tricíclicos/farmacocinética , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Morfina/farmacología , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Tiazepinas/metabolismo , Tiazepinas/farmacocinética
8.
J Med Chem ; 59(18): 8381-97, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27556704

RESUMEN

Natural products found in Mitragyna speciosa, commonly known as kratom, represent diverse scaffolds (indole, indolenine, and spiro pseudoindoxyl) with opioid activity, providing opportunities to better understand opioid pharmacology. Herein, we report the pharmacology and SAR studies both in vitro and in vivo of mitragynine pseudoindoxyl (3), an oxidative rearrangement product of the corynanthe alkaloid mitragynine. 3 and its corresponding corynantheidine analogs show promise as potent analgesics with a mechanism of action that includes mu opioid receptor agonism/delta opioid receptor antagonism. In vitro, 3 and its analogs were potent agonists in [(35)S]GTPγS assays at the mu opioid receptor but failed to recruit ß-arrestin-2, which is associated with opioid side effects. Additionally, 3 developed analgesic tolerance more slowly than morphine, showed limited physical dependence, respiratory depression, constipation, and displayed no reward or aversion in CPP/CPA assays, suggesting that analogs might represent a promising new generation of novel pain relievers.


Asunto(s)
Analgésicos Opioides/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Alcaloides de Triptamina Secologanina/farmacología , Arrestina beta 2/metabolismo , Analgésicos Opioides/química , Animales , Línea Celular , Humanos , Masculino , Ratones , Mitragyna/química , Simulación del Acoplamiento Molecular , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina Secologanina/química
9.
Synapse ; 70(10): 395-407, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27223691

RESUMEN

Buprenorphine has long been classified as a mu analgesic, although its high affinity for other opioid receptor classes and the orphanin FQ/nociceptin ORL1 receptor may contribute to its other actions. The current studies confirmed a mu mechanism for buprenorphine analgesia, implicating several subsets of mu receptor splice variants. Buprenorphine analgesia depended on the expression of both exon 1-associated traditional full length 7 transmembrane (7TM) and exon 11-associated truncated 6 transmembrane (6TM) MOR-1 variants. In genetic models, disruption of delta, kappa1 or ORL1 receptors had no impact on buprenorphine analgesia, while loss of the traditional 7TM MOR-1 variants in an exon 1 knockout (KO) mouse markedly lowered buprenorphine analgesia. Loss of the truncated 6TM variants in an exon 11 KO mouse totally eliminated buprenorphine analgesia. In distinction to analgesia, the inhibition of gastrointestinal transit and stimulation of locomotor activity were independent of truncated 6TM variants. Restoring expression of a 6TM variant with a lentivirus rescued buprenorphine analgesia in an exon 11 KO mouse that still expressed the 7TM variants. Despite a potent and robust stimulation of (35) S-GTPγS binding in MOR-1 expressing CHO cells, buprenorphine failed to recruit ß-arrestin-2 binding at doses as high as 10 µM. Buprenorphine was an antagonist in DOR-1 expressing cells and an inverse agonist in KOR-1 cells. Buprenorphine analgesia is complex and requires multiple mu receptor splice variant classes but other actions may involve alternative receptors.


Asunto(s)
Analgésicos Opioides/farmacología , Buprenorfina/farmacología , Nocicepción , Empalme del ARN , Receptores Opioides mu/genética , Animales , Células CHO , Cricetinae , Cricetulus , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores Opioides mu/metabolismo , Arrestina beta 2/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(13): 3663-8, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26976581

RESUMEN

The clinical management of severe pain depends heavily on opioids acting through mu opioid receptors encoded by the Oprm1 gene, which undergoes extensive alternative splicing. In addition to generating a series of prototypic seven transmembrane domain (7TM) G protein-coupled receptors (GPCRs), Oprm1 also produces a set of truncated splice variants containing only six transmembrane domains (6TM) through which selected opioids such as IBNtxA (3'-iodobenzoyl-6ß-naltrexamide) mediate a potent analgesia without many undesirable effects. Although morphine analgesia is independent of these 6TM mu receptor isoforms, we now show that the selective loss of the 6TM variants in a knockout model eliminates the analgesic actions of delta and kappa opioids and of α2-adrenergic compounds, but not cannabinoid, neurotensin, or muscarinic drugs. These observations were confirmed by using antisense paradigms. Despite their role in analgesia, loss of the 6TM variants were not involved with delta opioid-induced seizure activity, aversion to the kappa drug U50, 488H, or α2-mediated hypolocomotion. These observations support the existence of parallel opioid and nonopioid pain modulatory systems and highlight the ability to dissociate unwanted delta, kappa1, and α2 actions from analgesia.


Asunto(s)
Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Empalme Alternativo , Analgesia , Analgésicos Opioides/farmacología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Manejo del Dolor , Dimensión del Dolor , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Receptores Opioides mu/deficiencia
11.
Bioorg Med Chem Lett ; 25(8): 1761-1764, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25777268

RESUMEN

Trialkylstannanes are versatile precursors for chemical transformations, including radiolabeling with a variety of halogens, particularly iodine. In the present work a convenient, Pd-mediated stannylation method is presented that can be performed in an open flask. The method is selective for aryl iodides allowing selective stannylations in the presence of other halogen atoms. The reaction conditions are mild, making the method compatible with chemically sensitive bioactive compounds.


Asunto(s)
Yodo/química , Paladio/química , Animales , Encéfalo/metabolismo , Catálisis , Radioisótopos de Yodo/química , Marcaje Isotópico , Ratones , Piperidinas/química , Pirazoles/química
12.
J Pharmacol Exp Ther ; 350(3): 710-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24970924

RESUMEN

IBNtxA (3'-iodobenzoyl-6ß-naltrexamide) is a potent analgesic in mice lacking many traditional opioid side effects. In mice, it displays no respiratory depression, does not produce physical dependence with chronic administration, and shows no cross-tolerance to morphine. It has limited effects on gastrointestinal transit and shows no reward behavior. Biochemical studies indicate its actions are mediated through a set of µ-opioid receptor clone MOR-1 splice variants associated with exon 11 that lack exon 1 and contain only six transmembrane domains. Like the mouse and human, rats express exon 11-associated splice variants that also contain only six transmembrane domains, raising the question of whether IBNtxA would have a similar pharmacologic profile in rats. When given systemically, IBNtxA is a potent analgesic in rats, with an ED50 value of 0.89 mg/kg s.c., approximately 4-fold more potent than morphine. It shows no analgesic cross-tolerance in morphine-pelleted rats. IBNtxA displays no respiratory depression as measured by blood oxygen saturation. In contrast, oximetry shows that an equianalgesic dose of morphine lowers blood oxygen saturation values by 30%. IBNtxA binding is present in a number of brain regions, with the thalamus standing out with very high levels and the cerebellum with low levels. As in mice, IBNtxA is a potent analgesic in rats with a favorable pharmacologic profile and reduced side effects.


Asunto(s)
Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Naltrexona/análogos & derivados , Insuficiencia Respiratoria/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Naltrexona/metabolismo , Naltrexona/farmacología , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Unión Proteica/fisiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
13.
Eur J Med Chem ; 69: 786-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24103580

RESUMEN

Aminomorphinans are a relatively young class of opioid drugs among which substances of high in vitro efficacy and favorable in vivo action are found. We report the synthesis and pharmacological evaluation of novel 6ß-acylaminomorphinans. 6ß-Morphinamine and 6ß-codeinamine were stereoselectively synthesized by Mitsunobu reaction. The aminomorphinans were subsequently acylated with diversely substituted cinnamic acids. In vitro binding studies on cinnamoyl morphinamines showed moderate affinity for all opiate receptors with some selectivity for mu opioid receptors, while cinnamoyl codeinamines only showed affinity for mu opioid receptors. In vivo analgesia studies showed significant analgesic activity of 6ß-cinnamoylmorphinamine mediated by mu and delta receptors. The lead compound was found to be roughly equipotent to morphine (ED50 3.13 ± 1.09 mg/kg) but devoid of the dangerous side-effect respiratory depression, a major issue associated with traditional opioid therapy.


Asunto(s)
Analgésicos/farmacología , Morfinanos/farmacología , Umbral del Dolor/efectos de los fármacos , Analgésicos/síntesis química , Analgésicos/química , Animales , Relación Dosis-Respuesta a Droga , Calor , Ratones , Estructura Molecular , Morfinanos/síntesis química , Morfinanos/química , Morfina/efectos adversos , Morfina/farmacología , Antagonistas de Narcóticos , Dimensión del Dolor , Receptores Opioides/agonistas , Insuficiencia Respiratoria/inducido químicamente , Relación Estructura-Actividad , Factores de Tiempo
14.
Bioorg Med Chem Lett ; 23(15): 4347-50, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23796454

RESUMEN

Standard radioiodination methods lack site-selectivity and either mask charges (Bolton-Hunter) or involve oxidative reaction conditions (chloramine-T). Opioid peptides are very sensitive to certain structural modifications, making these labeling methods untenable. In our model opioid peptide, α-neoendorphin, we replaced a tyrosyl hydroxyl with an iodine, and in cell lines stably expressing mu, delta, or kappa opioid receptors, we saw no negative effects on binding. We then optimized a repurposed Sandmeyer reaction using copper(I) catalysts with non-redoxing/non-nucleophilic ligands, bringing the radiochemical yield up to around 30%, and site-selectively incorporated radioactive iodine into this position under non-oxidizing reaction conditions, which should be broadly compatible with most peptides. The (125)I- and (131)I-labeled versions of the compound bound with high affinity to opioid receptors in mouse brain homogenates, thus demonstrating the general utility of the labeling strategy and of the peptide for exploring opioid binding sites.


Asunto(s)
Endorfinas/metabolismo , Péptidos Opioides/química , Precursores de Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Encéfalo/metabolismo , Células CHO , Catálisis , Cobre/química , Cricetinae , Cricetulus , Endorfinas/química , Halogenación , Radioisótopos de Yodo/química , Ratones , Péptidos Opioides/síntesis química , Péptidos Opioides/metabolismo , Unión Proteica , Precursores de Proteínas/química , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...