Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 25, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167383

RESUMEN

Lipid nanodiscs have become a standard tool for studying membrane proteins, including using single particle cryo-electron microscopy (cryo-EM). We find that reconstituting the pentameric ligand-gated ion channel (pLGIC), Erwinia ligand-gated ion channel (ELIC), in different nanodiscs produces distinct structures by cryo-EM. The effect of the nanodisc on ELIC structure extends to the extracellular domain and agonist binding site. Additionally, molecular dynamic simulations indicate that nanodiscs of different size impact ELIC structure and that the nanodisc scaffold directly interacts with ELIC. These findings suggest that the nanodisc plays a crucial role in determining the structure of pLGICs, and that reconstitution of ion channels in larger nanodiscs may better approximate a lipid membrane environment.


Asunto(s)
Canales Iónicos Activados por Ligandos , Canales Iónicos Activados por Ligandos/química , Canales Iónicos Activados por Ligandos/metabolismo , Microscopía por Crioelectrón , Modelos Moleculares , Sitios de Unión , Lípidos
2.
Elife ; 112022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34982031

RESUMEN

Polyunsaturated fatty acids (PUFAs) inhibit pentameric ligand-gated ion channels (pLGICs) but the mechanism of inhibition is not well understood. The PUFA, docosahexaenoic acid (DHA), inhibits agonist responses of the pLGIC, ELIC, more effectively than palmitic acid, similar to the effects observed in the GABAA receptor and nicotinic acetylcholine receptor. Using photo-affinity labeling and coarse-grained molecular dynamics simulations, we identified two fatty acid binding sites in the outer transmembrane domain (TMD) of ELIC. Fatty acid binding to the photolabeled sites is selective for DHA over palmitic acid, and specific for an agonist-bound state. Hexadecyl-methanethiosulfonate modification of one of the two fatty acid binding sites in the outer TMD recapitulates the inhibitory effect of PUFAs in ELIC. The results demonstrate that DHA selectively binds to multiple sites in the outer TMD of ELIC, but that state-dependent binding to a single intrasubunit site mediates DHA inhibition of ELIC.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Canales Iónicos Activados por Ligandos/metabolismo , Sitios de Unión , Simulación de Dinámica Molecular , Dominios Proteicos
3.
Curr Opin Anaesthesiol ; 35(1): 53-59, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-34669613

RESUMEN

PURPOSE OF REVIEW: With an aging cardiac surgery population, prefrail and frail patients are becoming more common. Anesthesiologists will be faced with the decision of how best to provide care to frail patients. Identification, management, and outcomes in frail patients will be discussed in this review. RECENT FINDINGS: Frailty is associated with a variety of poor outcomes, such as increased hospital length of stay, medical resource utilization, readmission rates, and mortality. Prehabilitation may play a greater role in the management of frail cardiac surgery patients. SUMMARY: As frailty will likely only increase amongst cardiac surgery patients, it is important to develop multicenter trials to study management and treatment options. Until those studies are performed, the care of frail cardiac surgery patients may be best provided by high-volume surgical centers with expertise in the management of frail patients.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Fragilidad , Anciano , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Anciano Frágil , Fragilidad/complicaciones , Fragilidad/diagnóstico , Evaluación Geriátrica , Humanos , Tiempo de Internación , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/terapia , Factores de Riesgo
4.
J Cardiothorac Vasc Anesth ; 35(11): 3340-3349, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431271

RESUMEN

The sinuses of Valsalva are outpouchings in the aortic root just distal to the aortic valve that serve several physiologic functions. Aneurysm of this segment of the aorta is quite rare and infrequently encountered in clinical practice. Due to the rarity of sinus of Valsalva aneurysms, there is a lack of controlled trials and most of the literature consists of case reports and series. Here, the authors review the currently available literature to discuss the anatomy and normal function of the aortic root, as well as disease pathology and diagnostic imaging considerations. Using reported cases, the authors also will discuss considerations for cardiac anesthesiologists in the perioperative period.


Asunto(s)
Aneurisma de la Aorta , Seno Aórtico , Aorta , Aneurisma de la Aorta/diagnóstico por imagen , Aneurisma de la Aorta/cirugía , Válvula Aórtica , Humanos , Seno Aórtico/diagnóstico por imagen , Seno Aórtico/cirugía
5.
Front Physiol ; 12: 798102, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069257

RESUMEN

Lipids modulate the function of many ion channels, possibly through direct lipid-protein interactions. The recent outpouring of ion channel structures by cryo-EM has revealed many lipid binding sites. Whether these sites mediate lipid modulation of ion channel function is not firmly established in most cases. However, it is intriguing that many of these lipid binding sites are also known sites for other allosteric modulators or drugs, supporting the notion that lipids act as endogenous allosteric modulators through these sites. Here, we review such lipid-drug binding sites, focusing on pentameric ligand-gated ion channels and transient receptor potential channels. Notable examples include sites for phospholipids and sterols that are shared by anesthetics and vanilloids. We discuss some implications of lipid binding at these sites including the possibility that lipids can alter drug potency or that understanding protein-lipid interactions can guide drug design. Structures are only the first step toward understanding the mechanism of lipid modulation at these sites. Looking forward, we identify knowledge gaps in the field and approaches to address them. These include defining the effects of lipids on channel function in reconstituted systems using asymmetric membranes and measuring lipid binding affinities at specific sites using native mass spectrometry, fluorescence binding assays, and computational approaches.

6.
J Biol Chem ; 292(23): 9480-9492, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28420728

RESUMEN

General anesthetics exert their effects on the central nervous system by acting on ion channels, most notably pentameric ligand-gated ion channels. Although numerous studies have focused on pentameric ligand-gated ion channels, the details of anesthetic binding and channel modulation are still debated. A better understanding of the anesthetic mechanism of action is necessary for the development of safer and more efficacious drugs. Herein, we present a computational study identifying two anesthetic binding sites in the transmembrane domain of the Gloeobacter violaceus ligand-gated ion channel (GLIC) channel, characterize the putative binding pathway, and observe structural changes associated with channel function. Molecular simulations of desflurane reveal a binding pathway to GLIC via a membrane-embedded tunnel using an intrasubunit protein lumen as the conduit, an observation that explains the Meyer-Overton hypothesis, or why the lipophilicity of an anesthetic and its potency are generally proportional. Moreover, employing high concentrations of ligand led to the identification of a second transmembrane site (TM2) that inhibits dissociation of anesthetic from the TM1 site and is consistent with the high concentrations of anesthetics required to achieve clinical effects. Finally, asymmetric binding patterns of anesthetic to the channel were found to promote an iris-like conformational change that constricts and dehydrates the ion pore, creating a 13.5 kcal/mol barrier to ion translocation. Together with previous studies, the simulations presented herein demonstrate a novel anesthetic binding site in GLIC that is accessed through a membrane-embedded tunnel and interacts with a previously known site, resulting in conformational changes that produce a non-conductive state of the channel.


Asunto(s)
Anestésicos por Inhalación/química , Proteínas Bacterianas , Membrana Celular , Cianobacterias , Isoflurano/análogos & derivados , Canales Iónicos Activados por Ligandos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Cianobacterias/química , Cianobacterias/metabolismo , Desflurano , Transporte Iónico/fisiología , Isoflurano/química , Canales Iónicos Activados por Ligandos/química , Canales Iónicos Activados por Ligandos/metabolismo
7.
Biochim Biophys Acta ; 1858(10): 2290-2304, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27163493

RESUMEN

The cellular membrane constitutes the first element that encounters a wide variety of molecular species to which a cell might be exposed. Hosting a large number of structurally and functionally diverse proteins associated with this key metabolic compartment, the membrane not only directly controls the traffic of various molecules in and out of the cell, it also participates in such diverse and important processes as signal transduction and chemical processing of incoming molecular species. In this article, we present a number of cases where details of interaction of small molecular species such as drugs with the membrane, which are often experimentally inaccessible, have been studied using advanced molecular simulation techniques. We have selected systems in which partitioning of the small molecule with the membrane constitutes a key step for its final biological function, often binding to and interacting with a protein associated with the membrane. These examples demonstrate that membrane partitioning is not only important for the overall distribution of drugs and other small molecules into different compartments of the body, it may also play a key role in determining the efficiency and the mode of interaction of the drug with its target protein. This article is part of a Special Issue entitled: Biosimulations edited by Ilpo Vattulainen and Tomasz Róg.


Asunto(s)
Membrana Celular/química , Proteínas de la Membrana/química , Simulación de Dinámica Molecular , Anestésicos/farmacocinética , Anestésicos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Citocromo P-450 CYP3A/fisiología , Complejo IV de Transporte de Electrones/metabolismo , Oxígeno/metabolismo , Esteroides/farmacocinética
8.
Biochim Biophys Acta ; 1858(7 Pt B): 1573-83, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26940626

RESUMEN

Peripheral membrane proteins are structurally diverse proteins that are involved in fundamental cellular processes. Their activity of these proteins is frequently modulated through their interaction with cellular membranes, and as a result techniques to study the interfacial interaction between peripheral proteins and the membrane are in high demand. Due to the fluid nature of the membrane and the reversibility of protein-membrane interactions, the experimental study of these systems remains a challenging task. Molecular dynamics simulations offer a suitable approach to study protein-lipid interactions; however, the slow dynamics of the lipids often prevents sufficient sampling of specific membrane-protein interactions in atomistic simulations. To increase lipid dynamics while preserving the atomistic detail of protein-lipid interactions, in the highly mobile membrane-mimetic (HMMM) model the membrane core is replaced by an organic solvent, while short-tailed lipids provide a nearly complete representation of natural lipids at the organic solvent/water interface. Here, we present a brief introduction and a summary of recent applications of the HMMM to study different membrane proteins, complementing the experimental characterization of the presented systems, and we offer a perspective of future applications of the HMMM to study other classes of membrane proteins. This article is part of a Special Issue entitled: Membrane proteins edited by J.C. Gumbart and Sergei Noskov.


Asunto(s)
Membrana Dobles de Lípidos/química , Proteínas de la Membrana/química , Proteínas de la Membrana/ultraestructura , Modelos Químicos , Modelos Moleculares , Sitios de Unión , Simulación por Computador , Fluidez de la Membrana , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas/métodos
9.
J Membr Biol ; 248(3): 563-82, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25998378

RESUMEN

Biological membranes constitute a critical component in all living cells. In addition to providing a conducive environment to a wide range of cellular processes, including transport and signaling, mounting evidence has established active participation of specific lipids in modulating membrane protein function through various mechanisms. Understanding lipid-protein interactions underlying these mechanisms at a sufficiently high resolution has proven extremely challenging, partly due to the semi-fluid nature of the membrane. In order to address this challenge computationally, multiple methods have been developed, including an alternative membrane representation termed highly mobile membrane mimetic (HMMM) in which lateral lipid diffusion has been significantly enhanced without compromising atomic details. The model allows for efficient sampling of lipid-protein interactions at atomic resolution, thereby significantly enhancing the effectiveness of molecular dynamics simulations in capturing membrane-associated phenomena. In this review, after providing an overview of HMMM model development, we will describe briefly successful application of the model to study a variety of membrane processes, including lipid-dependent binding and insertion of peripheral proteins, the mechanism of phospholipid insertion into lipid bilayers, and characterization of optimal tilt angle of transmembrane helices. We conclude with practical recommendations for proper usage of the model in simulation studies of membrane processes.


Asunto(s)
Membrana Celular/ultraestructura , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Animales , Membrana Celular/química , Permeabilidad de la Membrana Celular , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lípidos de la Membrana/química , Proteínas de la Membrana/química , Solventes/química
10.
Biophys J ; 107(9): 2059-69, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25418091

RESUMEN

Experimental challenges associated with characterization of the membrane-bound form of talin have prevented us from understanding the molecular mechanism of its membrane-dependent integrin activation. Here, utilizing what we believe to be a novel membrane mimetic model, we present a reproducible model of membrane-bound talin observed across multiple independent simulations. We characterize both local and global membrane-induced structural transitions that successfully reconcile discrepancies between biochemical and structural studies and provide insight into how talin might modulate integrin function. Membrane binding of talin, captured in unbiased simulations, proceeds through three distinct steps: initial electrostatic recruitment of the F2 subdomain to anionic lipids via several basic residues; insertion of an initially buried, conserved hydrophobic anchor into the membrane; and association of the F3 subdomain with the membrane surface through a large, interdomain conformational change. These latter two steps, to our knowledge, have not been observed or described previously. Electrostatic analysis shows talin F2F3 to be highly polarized, with a highly positive underside, which we attribute to the initial electrostatic recruitment, and a negative top face, which can help orient the protein optimally with respect to the membrane, thereby reducing the number of unproductive membrane collision events.


Asunto(s)
Membranas Artificiales , Talina/metabolismo , Simulación por Computador , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Fosfatidilserinas/metabolismo , Conformación Proteica , Estabilidad Proteica , Electricidad Estática , Talina/química , Talina/genética
11.
Biophys J ; 107(9): 2112-21, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25418096

RESUMEN

The SNARE complex plays a vital role in vesicle fusion arising during neuronal exocytosis. Key components in the regulation of SNARE complex formation, and ultimately fusion, are the transmembrane and linker regions of the vesicle-associated protein, synaptobrevin. However, the membrane-embedded structure of synaptobrevin in its prefusion state, which determines its interaction with other SNARE proteins during fusion, is largely unknown. This study reports all-atom molecular-dynamics simulations of the prefusion configuration of synaptobrevin in a lipid bilayer, aimed at characterizing the insertion depth and the orientation of the protein in the membrane, as well as the nature of the amino acids involved in determining these properties. By characterizing the structural properties of both wild-type and mutant synaptobrevin, the effects of C-terminal additions on tilt and insertion depth of membrane-embedded synaptobrevin are determined. The simulations suggest a robust, highly tilted state for membrane-embedded synaptobrevin with a helical connection between the transmembrane and linker regions, leading to an apparently new characterization of structural elements in prefusion synaptobrevin and providing a framework for interpreting past mutation experiments.


Asunto(s)
Membrana Dobles de Lípidos/química , Proteínas R-SNARE/química , Secuencia de Aminoácidos , Simulación de Dinámica Molecular , Mutación , Fosfatidilcolinas/química , Proteínas R-SNARE/genética
12.
J Phys Chem B ; 118(42): 12075-86, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25303275

RESUMEN

While small molecules have been used to induce anesthesia in a clinical setting for well over a century, a detailed understanding of the molecular mechanism remains elusive. In this study, we utilize ab initio calculations to develop a novel set of CHARMM-compatible parameters for the ubiquitous modern anesthetics desflurane, isoflurane, sevoflurane, and propofol for use in molecular dynamics (MD) simulations. The parameters generated were rigorously tested against known experimental physicochemical properties including dipole moment, density, enthalpy of vaporization, and free energy of solvation. In all cases, the anesthetic parameters were able to reproduce experimental measurements, signifying the robustness and accuracy of the atomistic models developed. The models were then used to study the interaction of anesthetics with the membrane. Calculation of the potential of mean force for inserting the molecules into a POPC bilayer revealed a distinct energetic minimum of 4-5 kcal/mol relative to aqueous solution at the level of the glycerol backbone in the membrane. The location of this minimum within the membrane suggests that anesthetics partition to the membrane prior to binding their ion channel targets, giving context to the Meyer-Overton correlation. Moreover, MD simulations of these drugs in the membrane give rise to computed membrane structural parameters, including atomic distribution, deuterium order parameters, dipole potential, and lateral stress profile, that indicate partitioning of anesthetics into the membrane at the concentration range studied here, which does not appear to perturb the structural integrity of the lipid bilayer. These results signify that an indirect, membrane-mediated mechanism of channel modulation is unlikely.


Asunto(s)
Anestésicos Generales/química , Anestésicos Generales/metabolismo , Simulación por Computador , Simulación de Dinámica Molecular , Anestésicos Generales/farmacología , Membrana Celular/química , Membrana Celular/metabolismo , Canales Iónicos/metabolismo , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Conformación Molecular , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Reproducibilidad de los Resultados
13.
J Phys Chem B ; 118(6): 1481-92, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24451004

RESUMEN

Energetics of protein side chain partitioning between aqueous solution and cellular membranes is of fundamental importance for correctly capturing the membrane binding and specific protein-lipid interactions of peripheral membrane proteins. We recently reported a highly mobile membrane mimetic (HMMM) model that accelerates lipid dynamics by modeling the membrane interior partially as a fluid organic solvent while retaining a literal description of the lipid head groups and the beginning of the tails. While the HMMM has been successfully applied to study spontaneous insertion of a number of peripheral proteins into membranes, a quantitative characterization of the energetics of membrane-protein interactions in HMMM membranes has not been performed. We report here the free energy profiles for partitioning of 10 protein side chain analogues into a HMMM membrane. In the interfacial and headgroup regions of the membrane, the side chain free energy profiles show excellent agreement with profiles previously reported for conventional membranes with full-tail lipids. In regions where the organic solvent is prevalent, the increased dipole and fluidity of the solvent generally result in a less accurate description, most notably overstabilization of aromatic and polar amino acids. As an additional measure of the ability of the HMMM model to describe membrane-protein interactions, the water-to-membrane interface transfer energies were analyzed and found to be in agreement with the previously reported experimental and computational hydrophobicity scales. We discuss strengths and weaknesses of HMMM in describing protein-membrane interactions as well as further development of model membranes.


Asunto(s)
Aminoácidos/metabolismo , Membrana Celular/metabolismo , Simulación de Dinámica Molecular , Proteínas/química , Proteínas/metabolismo , Aminoácidos/química , Interacciones Hidrofóbicas e Hidrofílicas , Membranas Artificiales , Conformación Proteica , Termodinámica
14.
Biophys J ; 102(9): 2130-9, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22824277

RESUMEN

Characterizing atomic details of membrane binding of peripheral membrane proteins by molecular dynamics (MD) has been significantly hindered by the slow dynamics of membrane reorganization associated with the phenomena. To expedite lateral diffusion of lipid molecules without sacrificing the atomic details of such interactions, we have developed a novel membrane representation, to our knowledge, termed the highly mobile membrane-mimetic (HMMM) model to study binding and insertion of various molecular species into the membrane. The HMMM model takes advantage of an organic solvent layer to represent the hydrophobic core of the membrane and short-tailed phospholipids for the headgroup region. We demonstrate that using these components, bilayer structures are formed spontaneously and rapidly, regardless of the initial position and orientation of the lipids. In the HMMM membrane, lipid molecules exhibit one to two orders of magnitude enhancement in lateral diffusion. At the same time, the membrane atomic density profile of the headgroup region produced by the HMMM model is essentially identical to those obtained for full-membrane models, indicating the faithful representation of the membrane surface by the model. We demonstrate the efficiency of the model in capturing spontaneous binding and insertion of peripheral proteins by using the membrane anchor (γ-carboxyglutamic-acid-rich domain; GLA domain) of human coagulation factor VII as a test model. Achieving full insertion of the GLA domain consistently in 10 independent unbiased simulations within short simulation times clearly indicates the robustness of the HMMM model in capturing membrane association of peripheral proteins very efficiently and reproducibly. The HMMM model will provide significant improvements to the current all-atom models by accelerating lipid dynamics to examine protein-membrane interactions more efficiently.


Asunto(s)
Biomimética/métodos , Membrana Dobles de Lípidos/química , Fluidez de la Membrana , Proteínas de la Membrana/química , Proteínas de la Membrana/ultraestructura , Modelos Químicos , Modelos Moleculares , Simulación por Computador
15.
J Biol Chem ; 286(26): 23247-53, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21561861

RESUMEN

Many regulatory processes in biology involve reversible association of proteins with membranes. Clotting proteins bind to phosphatidylserine (PS) on cell surfaces, but a clear picture of this interaction has yet to emerge. We present a novel explanation for membrane binding by GLA domains of clotting proteins, supported by biochemical studies, solid-state NMR analyses, and molecular dynamics simulations. The model invokes a single "phospho-L-serine-specific" interaction and multiple "phosphate-specific" interactions. In the latter, the phosphates in phospholipids interact with tightly bound Ca(2+) in GLA domains. We show that phospholipids with any headgroup other than choline strongly synergize with PS to enhance factor X activation. We propose that phosphatidylcholine and sphingomyelin (the major external phospholipids of healthy cells) are anticoagulant primarily because their bulky choline headgroups sterically hinder access to their phosphates. Following cell damage or activation, exposed PS and phosphatidylethanolamine collaborate to bind GLA domains by providing phospho-L-serine-specific and phosphate-specific interactions, respectively.


Asunto(s)
Coagulación Sanguínea/fisiología , Calcio/metabolismo , Factor X/metabolismo , Fosfolípidos/metabolismo , Calcio/química , Factor X/química , Humanos , Resonancia Magnética Nuclear Biomolecular , Fosfolípidos/química
16.
J Phys Chem B ; 115(21): 7029-37, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21561114

RESUMEN

Membrane binding of peripheral proteins, mediated by specialized anchoring domains, is a crucial step for their biological function. Computational studies of membrane insertion, however, have proven challenging and largely inaccessible, due to the time scales required for the complete description of the process, mainly caused by the slow diffusion of the lipid molecules composing the membrane. Furthermore, in many cases, the nature of the membrane "anchor", i.e., the part of the protein that inserts into the membrane, is also unknown. Here, we address some of these issues by developing and employing a simplified representation of the membrane by a biphasic solvent model which we demonstrate can be used efficiently to capture and describe the process of hydrophobic insertion of membrane anchoring domains in all-atom molecular dynamics simulations. Applying the model, we have studied the insertion of the anchoring domain of a coagulation protein (the GLA domain of human protein C), starting from multiple initial configurations varying with regard to the initial orientation and height of the protein with respect to the membrane. In addition to efficiently and consistently identifying the "keel" region as the hydrophobic membrane anchor, within a few nanoseconds each configuration simulated showed a convergent height (2.20 ± 1.04 Å) and angle with respect to the interface normal (23.37 ± 12.48°). We demonstrate that the model can produce the same results as those obtained from a full representation of a membrane, in terms of both the depth of penetration and the orientation of the protein in the final membrane-bound form with an order of magnitude decrease in the required computational time compared to previous models, allowing for a more exhaustive search for the correct membrane-bound configuration.


Asunto(s)
Membranas Artificiales , Proteína C/química , Biocatálisis , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Proteína C/metabolismo , Conformación Proteica , Serina Proteasas/química , Serina Proteasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...