Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 180(4): 605-632, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32059777

RESUMEN

Despite advances in genetic and proteomic techniques, a complete portrait of the proteome and its complement of dynamic interactions and modifications remains a lofty, and as of yet, unrealized, objective. Specifically, traditional biological and analytical approaches have not been able to address key questions relating to the interactions of proteins with small molecules, including drugs, drug candidates, metabolites, or protein post-translational modifications (PTMs). Fortunately, chemists have bridged this experimental gap through the creation of bioorthogonal reactions. These reactions allow for the incorporation of chemical groups with highly selective reactivity into small molecules or protein modifications without perturbing their biological function, enabling the selective installation of an analysis tag for downstream investigations. The introduction of chemical strategies to parse and enrich subsets of the "functional" proteome has empowered mass spectrometry (MS)-based methods to delve more deeply and precisely into the biochemical state of cells and its perturbations by small molecules. In this Primer, we discuss how one of the most versatile bioorthogonal reactions, "click chemistry", has been exploited to overcome limitations of biological approaches to enable the selective marking and functional investigation of critical protein-small-molecule interactions and PTMs in native biological environments.


Asunto(s)
Química Clic/métodos , Proteómica/métodos , Descubrimiento de Drogas/métodos
2.
Cell ; 173(4): 815-817, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727668

RESUMEN

Matching genetically defined cancer states to drugs that specifically target these states is a principal goal of personalized oncology medicine. In this issue, McMillan et al. show how large-scale chemical screening coupled to deep molecular profiling can identify mechanistically diverse druggable vulnerabilities for genetic subtypes of lung cancers.


Asunto(s)
Perfilación de la Expresión Génica , Neoplasias Pulmonares , Humanos , Terapia Molecular Dirigida , Medicina de Precisión
3.
Cell ; 168(3): 527-541.e29, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28111073

RESUMEN

Advances in the synthesis and screening of small-molecule libraries have accelerated the discovery of chemical probes for studying biological processes. Still, only a small fraction of the human proteome has chemical ligands. Here, we describe a platform that marries fragment-based ligand discovery with quantitative chemical proteomics to map thousands of reversible small molecule-protein interactions directly in human cells, many of which can be site-specifically determined. We show that fragment hits can be advanced to furnish selective ligands that affect the activity of proteins heretofore lacking chemical probes. We further combine fragment-based chemical proteomics with phenotypic screening to identify small molecules that promote adipocyte differentiation by engaging the poorly characterized membrane protein PGRMC2. Fragment-based screening in human cells thus provides an extensive proteome-wide map of protein ligandability and facilitates the coordinated discovery of bioactive small molecules and their molecular targets.


Asunto(s)
Descubrimiento de Drogas/métodos , Proteómica/métodos , Adipocitos/citología , Diferenciación Celular , Cristalografía por Rayos X , Ensayos Analíticos de Alto Rendimiento , Humanos , Hidrolasas/química , Ligandos , Proteínas de la Membrana/antagonistas & inhibidores , Oxidorreductasas/química , Unión Proteica , Receptores de Progesterona/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas
4.
Nature ; 622(7983): 507-513, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37730997

RESUMEN

Marine-derived cyclic imine toxins, portimine A and portimine B, have attracted attention because of their chemical structure and notable anti-cancer therapeutic potential1-4. However, access to large quantities of these toxins is currently not feasible, and the molecular mechanism underlying their potent activity remains unknown until now. To address this, a scalable and concise synthesis of portimines is presented, which benefits from the logic used in the two-phase terpenoid synthesis5,6 along with other tactics such as exploiting ring-chain tautomerization and skeletal reorganization to minimize protecting group chemistry through self-protection. Notably, this total synthesis enabled a structural reassignment of portimine B and an in-depth functional evaluation of portimine A, revealing that it induces apoptosis selectively in human cancer cell lines with high potency and is efficacious in vivo in tumour-clearance models. Finally, practical access to the portimines and their analogues simplified the development of photoaffinity analogues, which were used in chemical proteomic experiments to identify a primary target of portimine A as the 60S ribosomal export protein NMD3.


Asunto(s)
Antineoplásicos , Técnicas de Química Sintética , Iminas , Compuestos de Espiro , Humanos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Iminas/síntesis química , Iminas/química , Iminas/farmacología , Neoplasias/tratamiento farmacológico , Proteómica , Ribosomas/metabolismo , Proteínas de Unión al ARN/metabolismo , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Relación Estructura-Actividad , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología
5.
Nat Chem Biol ; 20(7): 823-834, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38167919

RESUMEN

Photoaffinity probes are routinely utilized to identify proteins that interact with small molecules. However, despite this common usage, resolving the specific sites of these interactions remains a challenge. Here we developed a chemoproteomic workflow to determine precise protein binding sites of photoaffinity probes in cells. Deconvolution of features unique to probe-modified peptides, such as their tendency to produce chimeric spectra, facilitated the development of predictive models to confidently determine labeled sites. This yielded an expansive map of small-molecule binding sites on endogenous proteins and enabled the integration with multiplexed quantitation, increasing the throughput and dimensionality of experiments. Finally, using structural information, we characterized diverse binding sites across the proteome, providing direct evidence of their tractability to small molecules. Together, our findings reveal new knowledge for the analysis of photoaffinity probes and provide a robust method for high-resolution mapping of reversible small-molecule interactions en masse in native systems.


Asunto(s)
Etiquetas de Fotoafinidad , Bibliotecas de Moléculas Pequeñas , Sitios de Unión , Humanos , Etiquetas de Fotoafinidad/química , Bibliotecas de Moléculas Pequeñas/química , Unión Proteica , Proteómica/métodos , Proteoma/metabolismo , Proteínas/química , Proteínas/metabolismo , Péptidos/química , Péptidos/metabolismo
6.
Nat Chem Biol ; 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38191941

RESUMEN

SLC15A4 is an endolysosome-resident transporter linked with autoinflammation and autoimmunity. Specifically, SLC15A4 is critical for Toll-like receptors (TLRs) 7-9 as well as nucleotide-binding oligomerization domain-containing protein (NOD) signaling in several immune cell subsets. Notably, SLC15A4 is essential for the development of systemic lupus erythematosus in murine models and is associated with autoimmune conditions in humans. Despite its therapeutic potential, the availability of quality chemical probes targeting SLC15A4 functions is limited. In this study, we used an integrated chemical proteomics approach to develop a suite of chemical tools, including first-in-class functional inhibitors, for SLC15A4. We demonstrate that these inhibitors suppress SLC15A4-mediated endolysosomal TLR and NOD functions in a variety of human and mouse immune cells; we provide evidence of their ability to suppress inflammation in vivo and in clinical settings; and we provide insights into their mechanism of action. Our findings establish SLC15A4 as a druggable target for the treatment of autoimmune and autoinflammatory conditions.

7.
Nature ; 576(7785): 138-142, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31748741

RESUMEN

Haem is an essential prosthetic group of numerous proteins and a central signalling molecule in many physiologic processes1,2. The chemical reactivity of haem means that a network of intracellular chaperone proteins is required to avert the cytotoxic effects of free haem, but the constituents of such trafficking pathways are unknown3,4. Haem synthesis is completed in mitochondria, with ferrochelatase adding iron to protoporphyrin IX. How this vital but highly reactive metabolite is delivered from mitochondria to haemoproteins throughout the cell remains poorly defined3,4. Here we show that progesterone receptor membrane component 2 (PGRMC2) is required for delivery of labile, or signalling haem, to the nucleus. Deletion of PGMRC2 in brown fat, which has a high demand for haem, reduced labile haem in the nucleus and increased stability of the haem-responsive transcriptional repressors Rev-Erbα and BACH1. Ensuing alterations in gene expression caused severe mitochondrial defects that rendered adipose-specific PGRMC2-null mice unable to activate adaptive thermogenesis and prone to greater metabolic deterioration when fed a high-fat diet. By contrast, obese-diabetic mice treated with a small-molecule PGRMC2 activator showed substantial improvement of diabetic features. These studies uncover a role for PGRMC2 in intracellular haem transport, reveal the influence of adipose tissue haem dynamics on physiology and suggest that modulation of PGRMC2 may revert obesity-linked defects in adipocytes.


Asunto(s)
Adipocitos/metabolismo , Hemo/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/metabolismo , Animales , Homeostasis , Humanos , Espacio Intracelular/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Chaperonas Moleculares/metabolismo , Receptores de Progesterona/deficiencia , Receptores de Progesterona/genética , Transcripción Genética
8.
Proc Natl Acad Sci U S A ; 119(14): e2200544119, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35349343

RESUMEN

A function-impairing mutation (feeble) or genomic deletion of SLC15A4 abolishes responses of nucleic acid­sensing endosomal toll-like receptors (TLRs) and significantly reduces disease in mouse models of lupus. Here, we demonstrate disease reduction in homozygous and even heterozygous Slc15a4 feeble mutant BXSB male mice with a Tlr7 gene duplication. In contrast to SLC15A4, a function-impairing mutation of SLC15A3 did not diminish type I interferon (IFN-I) production by TLR-activated plasmacytoid dendritic cells (pDCs), indicating divergence of function between these homologous SLC15 family members. Trafficking to endolysosomes and function of SLC15A4 were dependent on the Adaptor protein 3 (AP-3) complex. Importantly, SLC15A4 was required for trafficking and colocalization of nucleic acid­sensing TLRs and their ligands to endolysosomes and the formation of the LAMP2+VAMP3+ hybrid compartment in which IFN-I production is initiated. Collectively, these findings define mechanistic processes by which SLC15A4 controls endosomal TLR function and suggest that pharmacologic intervention to curtail the function of this transporter may be a means to treat lupus and other endosomal TLR-dependent diseases.


Asunto(s)
Ácidos Nucleicos , Animales , Endosomas/metabolismo , Ligandos , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Receptores Toll-Like/metabolismo
10.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33926964

RESUMEN

Aberrant Ras signaling is linked to a wide spectrum of hyperproliferative diseases, and components of the signaling pathway, including Ras, have been the subject of intense and ongoing drug discovery efforts. The cellular activity of Ras is modulated by its association with the guanine nucleotide exchange factor Son of sevenless (Sos), and the high-resolution crystal structure of the Ras-Sos complex provides a basis for the rational design of orthosteric Ras ligands. We constructed a synthetic Sos protein mimic that engages the wild-type and oncogenic forms of nucleotide-bound Ras and modulates downstream kinase signaling. The Sos mimic was designed to capture the conformation of the Sos helix-loop-helix motif that makes critical contacts with Ras in its switch region. Chemoproteomic studies illustrate that the proteomimetic engages Ras and other cellular GTPases. The synthetic proteomimetic resists proteolytic degradation and enters cells through macropinocytosis. As such, it is selectively toxic to cancer cells with up-regulated macropinocytosis, including those that feature oncogenic Ras mutations.


Asunto(s)
Complejos Multiproteicos/ultraestructura , Conformación Proteica , Proteína Son Of Sevenless Drosofila/ultraestructura , Proteínas ras/ultraestructura , Animales , Biomimética , Cristalografía por Rayos X , Descubrimiento de Drogas , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/ultraestructura , Células HCT116 , Secuencias Hélice-Asa-Hélice/genética , Humanos , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Proteoma/genética , Transducción de Señal/genética , Proteína Son Of Sevenless Drosofila/química , Proteína Son Of Sevenless Drosofila/genética , Proteínas ras/química , Proteínas ras/genética
11.
Isr J Chem ; 63(3-4)2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38213795

RESUMEN

Chemical probes are invaluable tools to investigate biological processes and can serve as lead molecules for the development of new therapies. However, despite their utility, only a fraction of human proteins have selective chemical probes, and more generally, our knowledge of the "chemically-tractable" proteome is limited, leaving many potential therapeutic targets unexploited. To help address these challenges, powerful chemical proteomic approaches have recently been developed to globally survey the ability of proteins to bind small molecules (i. e., ligandability) directly in native systems. In this review, we discuss the utility of such approaches, with a focus on the integration of chemoproteomic methods with fragment-based ligand discovery (FBLD), to facilitate the broad mapping of the ligandable proteome while also providing starting points for progression into lead chemical probes.

12.
Proc Natl Acad Sci U S A ; 117(44): 27329-27338, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33067390

RESUMEN

Galectin-3 is a glycan-binding protein (GBP) that binds ß-galactoside glycan structures to orchestrate a variety of important biological events, including the activation of hepatic stellate cells and regulation of immune responses. While the requisite glycan epitopes needed to bind galectin-3 have long been elucidated, the cellular glycoproteins that bear these glycan signatures remain unknown. Given the importance of the three-dimensional (3D) arrangement of glycans in dictating GBP interactions, strategies that allow the identification of GBP receptors in live cells, where the native glycan presentation and glycoprotein expression are preserved, have significant advantages over static and artificial systems. Here we describe the integration of a proximity labeling method and quantitative mass spectrometry to map the glycan and glycoprotein interactors for galectin-3 in live human hepatic stellate cells and peripheral blood mononuclear cells. Understanding the identity of the glycoproteins and defining the structures of the glycans will empower efforts to design and develop selective therapeutics to mitigate galectin-3-mediated biological events.


Asunto(s)
Galectina 3/metabolismo , Polisacáridos/metabolismo , Técnicas de Cultivo de Célula , Galectina 3/fisiología , Galectinas/química , Glicoproteínas/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Polisacáridos/fisiología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/fisiología , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 117(52): 33197-33203, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33318191

RESUMEN

RNAs have important functions that are dictated by their structure. Indeed, small molecules that interact with RNA structures can perturb function, serving as chemical probes and lead medicines. Here we describe the development of a fragment-based approach to discover and optimize bioactive small molecules targeting RNA. We extended the target validation method chemical cross-linking and isolation by pull-down (Chem-CLIP) to identify and map the binding sites of low molecular weight fragments that engage RNA or Chem-CLIP fragment mapping (Chem-CLIP-Frag-Map). Using Chem-CLIP-Frag-Map, we identified several fragments that bind the precursor to oncogenic microRNA-21 (pre-miR-21). Assembly of these fragments provided a specific bioactive compound with improved potency that inhibits pre-miR-21 processing, reducing mature miR-21 levels. The compound exerted selective effects on the transcriptome and selectively mitigated a miR-21-associated invasive phenotype in triple-negative breast cancer cells. The Chem-CLIP-Frag-Map approach should prove general to expedite the identification and optimization of small molecules that bind RNA targets.


Asunto(s)
Antineoplásicos/química , Descubrimiento de Drogas/métodos , MicroARNs/química , Bibliotecas de Moléculas Pequeñas/química , Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Ligandos , MicroARNs/metabolismo , Simulación del Acoplamiento Molecular , Motivos de Nucleótidos , Bibliotecas de Moléculas Pequeñas/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo
14.
J Am Chem Soc ; 144(33): 15013-15019, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-35960875

RESUMEN

Heme is an essential cofactor for many human proteins as well as the primary transporter of oxygen in blood. Recent studies have also established heme as a signaling molecule, imparting its effects through binding with protein partners rather than through reactivity of its metal center. However, the comprehensive annotation of such heme-binding proteins in the human proteome remains incomplete. Here, we describe a strategy which utilizes a heme-based photoaffinity probe integrated with quantitative proteomics to map heme-protein interactions across the proteome. In these studies, we identified 350+ unique heme-protein interactions, the vast majority of which were heretofore unknown and consist of targets from diverse functional classes, including transporters, receptors, enzymes, transcription factors, and chaperones. Among these proteins is the immune-related interleukin receptor-associated kinase 1 (IRAK1), where we provide preliminary evidence that heme agonizes its catalytic activity. Our findings should improve the current understanding of heme's regulation as well as its signaling functions and facilitate new insights of its roles in human disease.


Asunto(s)
Hemo , Proteómica , Proteínas Portadoras/metabolismo , Hemo/química , Humanos , Unión Proteica , Proteoma/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
15.
J Am Chem Soc ; 143(40): 16700-16708, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34592107

RESUMEN

Protein acetylation is a central event in orchestrating diverse cellular processes. However, current strategies to investigate protein acetylation in cells are often nonspecific or lack temporal and magnitude control. Here, we developed an acetylation tagging system, AceTAG, to induce acetylation of targeted proteins. The AceTAG system utilizes bifunctional molecules to direct the lysine acetyltransferase p300/CBP to proteins fused with the small protein tag FKBP12F36V, resulting in their induced acetylation. Using AceTAG, we induced targeted acetylation of a diverse array of proteins in cells, specifically histone H3.3, the NF-κB subunit p65/RelA, and the tumor suppressor p53. We demonstrate that targeted acetylation with the AceTAG system is rapid, selective, reversible and can be controlled in a dose-dependent fashion. AceTAG represents a useful strategy to modulate protein acetylation and should enable the exploration of targeted acetylation in basic biological and therapeutic contexts.


Asunto(s)
Factor de Transcripción ReIA
17.
Methods Enzymol ; 681: 287-323, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36764762

RESUMEN

Protein acetylation is a vital biological process that regulates myriad cellular events. Despite its profound effects on protein function, there are limited research tools to dynamically and selectively regulate protein acetylation. To address this, we developed an acetylation tagging system, called AceTAG, to target proteins for chemically induced acetylation directly in live cells. AceTAG uses heterobifunctional molecules composed of a ligand for the lysine acetyltransferase p300/CBP and a FKBP12F36V ligand. Target proteins are genetically tagged with FKBP12F36V and brought in proximity with p300/CBP by AceTAG molecules to subsequently undergo protein-specific acetylation. Targeted acetylation of proteins in cells using AceTAG is selective, rapid, and can be modulated in a dose-dependent fashion, enabling controlled investigations of acetylated protein targets directly in cells. In this protocol, we focus on (1) generation of AceTAG constructs and cell lines, (2) in vitro characterization of AceTAG mediated ternary complex formation and cellular target engagement studies; and (3) in situ characterization of AceTAG induced acetylation of targeted proteins by immunoblotting and quantitative proteomics. The robust procedures described herein should enable the use of AceTAG to explore the roles of acetylation for a variety of protein targets.


Asunto(s)
Proteína 1A de Unión a Tacrolimus , Acetilación , Ligandos , Línea Celular
18.
Sci Transl Med ; 15(719): eadh1892, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37878674

RESUMEN

Programmed cell death protein 1 (PD-1) immune checkpoint blockade therapy has revolutionized cancer treatment. Although PD-1 blockade is effective in a subset of patients with cancer, many fail to respond because of either primary or acquired resistance. Thus, next-generation strategies are needed to expand the depth and breadth of clinical responses. Toward this end, we designed a human primary T cell phenotypic high-throughput screening strategy to identify small molecules with distinct and complementary mechanisms of action to PD-1 checkpoint blockade. Through these efforts, we selected and optimized a chemical series that showed robust potentiation of T cell activation and combinatorial activity with αPD-1 blockade. Target identification was facilitated by chemical proteomic profiling with a lipid-based photoaffinity probe, which displayed enhanced binding to diacylglycerol kinase α (DGKα) in the presence of the active compound, a phenomenon that correlated with the translocation of DGKα to the plasma membrane. We further found that optimized leads within this chemical series were potent and selective inhibitors of both DGKα and DGKζ, lipid kinases that constitute an intracellular T cell checkpoint that blunts T cell signaling through diacylglycerol metabolism. We show that dual DGKα/ζ inhibition amplified suboptimal T cell receptor signaling mediated by low-affinity antigen presentation and low major histocompatibility complex class I expression on tumor cells, both hallmarks of resistance to PD-1 blockade. In addition, DGKα/ζ inhibitors combined with αPD-1 therapy to elicit robust tumor regression in syngeneic mouse tumor models. Together, these findings support targeting DGKα/ζ as a next-generation T cell immune checkpoint strategy.


Asunto(s)
Neoplasias , Receptor de Muerte Celular Programada 1 , Ratones , Animales , Humanos , Receptor de Muerte Celular Programada 1/metabolismo , Proteómica , Diacilglicerol Quinasa/metabolismo , Linfocitos T , Lípidos
19.
RSC Chem Biol ; 3(12): 1369-1374, 2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36544572

RESUMEN

Human milk oligosaccharides (HMOs) are a family of unconjugated soluble glycans found in human breast milk that exhibit a myriad of biological activity. While recent studies have uncovered numerous biological functions for HMOs (antimicrobial, anti-inflammatory & probiotic properties), the receptors and protein binding partners involved in these processes are not well characterized. This can be attributed largely in part to the low affinity and transient nature of soluble glycan-protein interactions, precluding the use of traditional characterization techniques to survey binding partners in live cells. Here, we present the use of synthetic photoactivatable HMO probes to capture, enrich and identify HMO protein targets in live cells using mass spectrometry-based chemoproteomics. Following initial validation studies using purified lectins, we profiled the targets of HMO probes in live mouse macrophages. Using this strategy, we mapped hundreds of HMO binding partners across multiple cellular compartments, including many known glycan-binding proteins as well as numerous proteins previously not known to bind glycans. We expect our findings to inform future investigations of the diverse roles of how HMOs may regulate protein function.

20.
Cell Chem Biol ; 28(3): 371-393, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33577749

RESUMEN

The ID of disease-modifying, chemically accessible targets remains a central priority of modern therapeutic discovery. The phenotypic screening of small-molecule libraries not only represents an attractive approach to identify compounds that may serve as drug leads but also serves as an opportunity to uncover compounds with novel mechanisms of action (MoAs). However, a major bottleneck of phenotypic screens continues to be the ID of pharmacologically relevant target(s) for compounds of interest. The field of chemoproteomics aims to map proteome-wide small-molecule interactions in complex, native systems, and has proved a key technology to unravel the protein targets of pharmacological modulators. In this review, we discuss the application of modern chemoproteomic methods to identify protein targets of phenotypic screening hits and investigate MoAs, with a specific focus on the development of chemoproteomic-enabled compound libraries to streamline target discovery.


Asunto(s)
Preparaciones Farmacéuticas/química , Proteómica , Bibliotecas de Moléculas Pequeñas/química , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Humanos , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA