Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cell ; 81(22): 4605-4621.e11, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34582793

RESUMEN

G-protein-coupled receptors (GPCRs), also known as seven transmembrane receptors (7TMRs), typically interact with two distinct signal-transducers, i.e., G proteins and ß-arrestins (ßarrs). Interestingly, there are some non-canonical 7TMRs that lack G protein coupling but interact with ßarrs, although an understanding of their transducer coupling preference, downstream signaling, and structural mechanism remains elusive. Here, we characterize two such non-canonical 7TMRs, namely, the decoy D6 receptor (D6R) and the complement C5a receptor subtype 2 (C5aR2), in parallel with their canonical GPCR counterparts. We discover that D6R and C5aR2 efficiently couple to ßarrs, exhibit distinct engagement of GPCR kinases (GRKs), and activate non-canonical downstream signaling pathways. We also observe that ßarrs adopt distinct conformations for D6R and C5aR2, compared to their canonical GPCR counterparts, in response to common natural agonists. Our study establishes D6R and C5aR2 as ßarr-coupled 7TMRs and provides key insights into their regulation and signaling with direct implication for biased agonism.


Asunto(s)
Membrana Celular/metabolismo , Conformación Proteica , Transducción de Señal , beta-Arrestinas/química , Animales , Proteínas de Unión al GTP/química , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Transporte de Proteínas , Receptor de Anafilatoxina C5a/metabolismo
2.
Nature ; 583(7818): 862-866, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32555462

RESUMEN

The ß1-adrenoceptor (ß1AR) is a G-protein-coupled receptor (GPCR) that couples1 to the heterotrimeric G protein Gs. G-protein-mediated signalling is terminated by phosphorylation of the C terminus of the receptor by GPCR kinases (GRKs) and by coupling of ß-arrestin 1 (ßarr1, also known as arrestin 2), which displaces Gs and induces signalling through the MAP kinase pathway2. The ability of synthetic agonists to induce signalling preferentially through either G proteins or arrestins-known as biased agonism3-is important in drug development, because the therapeutic effect may arise from only one signalling cascade, whereas the other pathway may mediate undesirable side effects4. To understand the molecular basis for arrestin coupling, here we determined the cryo-electron microscopy structure of the ß1AR-ßarr1 complex in lipid nanodiscs bound to the biased agonist formoterol5, and the crystal structure of formoterol-bound ß1AR coupled to the G-protein-mimetic nanobody6 Nb80. ßarr1 couples to ß1AR in a manner distinct to that7 of Gs coupling to ß2AR-the finger loop of ßarr1 occupies a narrower cleft on the intracellular surface, and is closer to transmembrane helix H7 of the receptor when compared with the C-terminal α5 helix of Gs. The conformation of the finger loop in ßarr1 is different from that adopted by the finger loop of visual arrestin when it couples to rhodopsin8. ß1AR coupled to ßarr1 shows considerable differences in structure compared with ß1AR coupled to Nb80, including an inward movement of extracellular loop 3 and the cytoplasmic ends of H5 and H6. We observe weakened interactions between formoterol and two serine residues in H5 at the orthosteric binding site of ß1AR, and find that formoterol has a lower affinity for the ß1AR-ßarr1 complex than for the ß1AR-Gs complex. The structural differences between these complexes of ß1AR provide a foundation for the design of small molecules that could bias signalling in the ß-adrenoceptors.


Asunto(s)
Microscopía por Crioelectrón , Fumarato de Formoterol/química , Fumarato de Formoterol/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/ultraestructura , beta-Arrestina 1/química , beta-Arrestina 1/ultraestructura , Secuencia de Aminoácidos , Animales , Sitios de Unión , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/ultraestructura , Células HEK293 , Humanos , Modelos Moleculares , Complejos Multiproteicos , Receptores Adrenérgicos beta 1/metabolismo , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/metabolismo , Anticuerpos de Cadena Única/ultraestructura , Pez Cebra , beta-Arrestina 1/metabolismo
3.
EMBO Rep ; 21(9): e49886, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32715625

RESUMEN

ß-arrestins (ßarrs) are key regulators of G protein-coupled receptor (GPCR) signaling and trafficking, and their knockdown typically leads to a decrease in agonist-induced ERK1/2 MAP kinase activation. Interestingly, for some GPCRs, knockdown of ßarr1 augments agonist-induced ERK1/2 phosphorylation although a mechanistic basis for this intriguing phenomenon is unclear. Here, we use selected GPCRs to explore a possible correlation between the spatial positioning of receptor phosphorylation sites and the contribution of ßarr1 in ERK1/2 activation. We discover that engineering a spatially positioned double-phosphorylation-site cluster in the bradykinin receptor (B2 R), analogous to that present in the vasopressin receptor (V2 R), reverses the contribution of ßarr1 in ERK1/2 activation from inhibitory to promotive. An intrabody sensor suggests a conformational mechanism for this role reversal of ßarr1, and molecular dynamics simulation reveals a bifurcated salt bridge between this double-phosphorylation site cluster and Lys294 in the lariat loop of ßarr1, which directs the orientation of the lariat loop. Our findings provide novel insights into the opposite roles of ßarr1 in ERK1/2 activation for different GPCRs with a direct relevance to biased agonism and novel therapeutics.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Receptores Acoplados a Proteínas G , Células HEK293 , Humanos , Fosforilación , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
4.
J Biol Chem ; 295(30): 10153-10167, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32439801

RESUMEN

Agonist stimulation of G-protein-coupled receptors (GPCRs) typically leads to phosphorylation of GPCRs and binding to multifunctional proteins called ß-arrestins (ßarrs). The GPCR-ßarr interaction critically contributes to GPCR desensitization, endocytosis, and downstream signaling, and GPCR-ßarr complex formation can be used as a generic readout of GPCR and ßarr activation. Although several methods are currently available to monitor GPCR-ßarr interactions, additional sensors to visualize them may expand the toolbox and complement existing methods. We have previously described antibody fragments (FABs) that recognize activated ßarr1 upon its interaction with the vasopressin V2 receptor C-terminal phosphopeptide (V2Rpp). Here, we demonstrate that these FABs efficiently report the formation of a GPCR-ßarr1 complex for a broad set of chimeric GPCRs harboring the V2R C terminus. We adapted these FABs to an intrabody format by converting them to single-chain variable fragments and used them to monitor the localization and trafficking of ßarr1 in live cells. We observed that upon agonist simulation of cells expressing chimeric GPCRs, these intrabodies first translocate to the cell surface, followed by trafficking into intracellular vesicles. The translocation pattern of intrabodies mirrored that of ßarr1, and the intrabodies co-localized with ßarr1 at the cell surface and in intracellular vesicles. Interestingly, we discovered that intrabody sensors can also report ßarr1 recruitment and trafficking for several unmodified GPCRs. Our characterization of intrabody sensors for ßarr1 recruitment and trafficking expands currently available approaches to visualize GPCR-ßarr1 binding, which may help decipher additional aspects of GPCR signaling and regulation.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/metabolismo , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Transporte de Proteínas , Receptores Acoplados a Proteínas G/genética , beta-Arrestina 1/genética
5.
Protein Expr Purif ; 171: 105617, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32145391

RESUMEN

Chemokine receptors form a major sub-family of G protein-coupled receptors (GPCRs) and they are involved in a number of cellular and physiological processes related to our immune response and regulation. A better structural understanding of ligand-binding, activation, signaling and regulation of chemokine receptors is very important to design potentially therapeutic interventions for human disorders arising from aberrant chemokine signaling. One of the key limitations in probing the structural details of chemokine receptors is the availability of large amounts of purified, homogenous and fully functional chemokine ligands, and the commercially available products, are not affordable for in-depth structural studies. Moreover, production of uniformly isotope-labeled chemokines, for example, suitable for NMR-based structural investigation, also remains challenging. Here, we have designed a streamlined approach to express and purify the human chemokine CCL7 as well as its 15N-, 15N/13C-, 2H/15N/13C- isotope-labeled derivatives, at milligram levels using E. coli expression system. Purified CCL7 not only maintains a well-folded three-dimensional structure as analyzed using circular dichroism and 1H/15N NMR but it also induces coupling of heterotrimeric G-proteins and ß-arrestins for selected chemokine receptors in cellular system. We compared cAMP response induced by histidine tagged CCL7 and native CCL7 and found that modification of the N-terminus of CCL7 compromises its functionality. Our strategy presented here may be applicable to other chemokines and therefore, provide a potentially generic and cost-effective approach to produce chemokines in large amounts for functional and structural studies.


Asunto(s)
Quimiocina CCL7 , Receptores de Quimiocina , Quimiocina CCL7/biosíntesis , Quimiocina CCL7/química , Quimiocina CCL7/genética , Quimiocina CCL7/aislamiento & purificación , Células HEK293 , Humanos , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
6.
Artículo en Inglés | MEDLINE | ID: mdl-38831575

RESUMEN

Androgen (AR) signaling is the main signaling for the development of the prostate and its normal functioning. AR is highly specific for testosterone and dihydrotestosterone, significantly contributing to prostate development, physiology, and cancer. All these receptors have emerged as crucial therapeutic targets for PCa. In the year 1966, the Noble prize was awarded to Huggins and Hodge for their groundbreaking discovery of AR. As it is a pioneer transcription factor, it belongs to the steroid hormone receptor family and consists of domains, including DNA binding domain (DBD), hormone response elements (HRE), C-terminal ligand binding domain (LBD), and N-terminal regulatory domains. Structural variations in AR, such as AR gene amplification, LBD mutations, alternative splicing of exons, hypermethylation of AR, and co- regulators, are major contributors to PCa. It's signaling is crucial for the development and functioning of the prostate gland, with the AR being the key player. The specificity of AR for testosterone and dihydrotestosterone is important in prostate physiology. However, when it is dysregulated, AR contributes significantly to PCa. However, the structural variations in AR, such as gene amplification, mutations, alternative splicing, and epigenetic modifications, drive the PCa progression. Therefore, understanding AR function and dysregulation is essential for developing effective therapeutic strategies. Thus, the aim of this review was to examine how AR was initially pivotal for prostate development and how it turned out to show both positive and detrimental implications for the prostate.

7.
Nat Commun ; 14(1): 4808, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37558722

RESUMEN

Chemokine receptors constitute an important subfamily of G protein-coupled receptors (GPCRs), and they are critically involved in a broad range of immune response mechanisms. Ligand promiscuity among these receptors makes them an interesting target to explore multiple aspects of biased agonism. Here, we comprehensively characterize two chemokine receptors namely, CXCR4 and CXCR7, in terms of their transducer-coupling and downstream signaling upon their stimulation by a common chemokine agonist, CXCL12, and a small molecule agonist, VUF11207. We observe that CXCR7 lacks G-protein-coupling while maintaining robust ßarr recruitment with a major contribution of GRK5/6. On the other hand, CXCR4 displays robust G-protein activation as expected but exhibits significantly reduced ßarr-coupling compared to CXCR7. These two receptors induce distinct ßarr conformations even when activated by the same agonist, and CXCR7, unlike CXCR4, fails to activate ERK1/2 MAP kinase. We also identify a key contribution of a single phosphorylation site in CXCR7 for ßarr recruitment and endosomal localization. Our study provides molecular insights into intrinsic-bias encoded in the CXCR4-CXCR7 system with broad implications for drug discovery.


Asunto(s)
Receptores CXCR , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Proteínas de Unión al GTP , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quimiocina CXCL12/metabolismo
8.
Methods Cell Biol ; 169: 267-278, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35623705

RESUMEN

Agonist-induced interaction of ß-arrestins with GPCRs is critically involved in downstream signaling and regulation. This interaction is associated with activation and major conformational changes in ß-arrestins. Although there are some assays available to monitor the conformational changes in ß-arrestins in cellular context, additional sensors to report ß-arrestin activation, preferably with high-throughput capability, are likely to be useful considering the structural and functional diversity in GPCR-ß-arrestin complexes. We have recently developed an intrabody-based sensor as an integrated approach to monitor GPCR-ß-arrestin interaction and conformational change, and generated a luminescence-based reporter using NanoBiT complementation technology. This sensor is derived from a synthetic antibody fragment referred to as Fab30 that selectively recognizes activated and receptor-bound conformation of ß-arrestin1. Here, we present a step-by-step protocol to employ this intrabody sensor to measure the interaction and conformational activation of ß-arrestin1 upon agonist-stimulation of a prototypical GPCR, the complement C5a receptor (C5aR1). This protocol is potentially applicable to other GPCRs and may also be leveraged to deduce qualitative differences in ß-arrestin1 conformations induced by different ligands and receptor mutants.


Asunto(s)
Bioensayo , Luminiscencia , Conformación Molecular , beta-Arrestina 1 , beta-Arrestinas
9.
Nat Commun ; 13(1): 4634, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35941121

RESUMEN

Agonist-induced phosphorylation of G protein-coupled receptors (GPCRs) is a primary determinant of ß-arrestin (ßarr) recruitment and trafficking. For several GPCRs such as the vasopressin receptor subtype 2 (V2R), agonist-stimulation first drives the translocation of ßarrs to the plasma membrane, followed by endosomal trafficking, which is generally considered to be orchestrated by multiple phosphorylation sites. We have previously shown that mutation of a single phosphorylation site in the V2R (i.e., V2RT360A) results in near-complete loss of ßarr translocation to endosomes despite robust recruitment to the plasma membrane, and compromised ERK1/2 activation. Here, we discover that a synthetic intrabody (Ib30), which selectively recognizes activated ßarr1, efficiently rescues the endosomal trafficking of ßarr1 and ERK1/2 activation for V2RT360A. Molecular dynamics simulations reveal that Ib30 enriches active-like ßarr1 conformation with respect to the inter-domain rotation, and cellular assays demonstrate that it also enhances ßarr1-ß2-adaptin interaction. Our data provide an experimental framework to positively modulate the receptor-transducer-effector axis for GPCRs using intrabodies, which can be potentially integrated in the paradigm of GPCR-targeted drug discovery.


Asunto(s)
Receptores Acoplados a Proteínas G , Transducción de Señal , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , Arrestina beta 2/metabolismo , beta-Arrestinas/metabolismo
10.
Sci Signal ; 14(677)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824179

RESUMEN

The opioid crisis represents a major worldwide public health crisis that has accelerated the search for safer and more effective opioids. Over the past few years, the identification of biased opioid ligands capable of eliciting selective functional responses has provided an alternative avenue to develop novel therapeutics without the side effects of current opioid medications. However, whether biased agonism or other pharmacological properties, such as partial agonism (or low efficacy), account for the therapeutic benefits remains questionable. Here, we provide a summary of the current status of biased opioid ligands that target the µ- and κ-opioid receptors and highlight advances in preclinical and clinical trials of some of these ligands. We also discuss an example of structure-based biased ligand discovery at the µ-opioid receptor, an approach that could revolutionize drug discovery at opioid and other receptors. Last, we briefly discuss caveats and future directions for this important area of research.


Asunto(s)
Receptores Opioides mu , Receptores Opioides , Analgésicos Opioides , Ligandos , Receptores Opioides kappa
11.
Nat Commun ; 12(1): 7158, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34887409

RESUMEN

ß-arrestins (ßarrs) play multifaceted roles in the function of G protein-coupled receptors (GPCRs). ßarrs typically interact with phosphorylated C-terminal tail (C tail) and transmembrane core (TM core) of GPCRs. However, the effects of the C tail- and TM core-mediated interactions on the conformational activation of ßarrs have remained elusive. Here, we show the conformational changes for ßarr activation upon the C tail- and TM core-mediated interactions with a prototypical GPCR by nuclear magnetic resonance (NMR) spectroscopy. Our NMR analyses demonstrated that while the C tail-mediated interaction alone induces partial activation, in which ßarr exists in equilibrium between basal and activated conformations, the TM core- and the C tail-mediated interactions together completely shift the equilibrium toward the activated conformation. The conformation-selective antibody, Fab30, promotes partially activated ßarr into the activated-like conformation. This plasticity of ßarr conformation in complex with GPCRs engaged in different binding modes may explain the multifunctionality of ßarrs.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/química , beta-Arrestina 1/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Unión Proteica , Conformación Proteica en Lámina beta , Dominios Proteicos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , beta-Arrestina 1/genética
12.
Adv Cancer Res ; 145: 139-156, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32089163

RESUMEN

ß-Arrestins (ßarrs) are multifunctional intracellular proteins with an ability to directly interact with a large number of cellular partners including the G protein-coupled receptors (GPCRs). ßarrs contribute to multiple aspects of GPCR signaling, trafficking and downregulation. Considering the central involvement of GPCR signaling in the onset and progression of diverse types of cancers, ßarrs have also emerged as key players in the context of investigating cancer phenotypes, and as potential therapeutic targets. In this chapter, we first provide a brief account of structure and function of ßarrs and then highlight recent discoveries unfolding novel functional attributes of ßarrs in breast cancer. We also underscore the recent paradigms of modulating ßarr functions in cellular context and potential therapeutic opportunities going forward.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Terapia Molecular Dirigida , beta-Arrestinas/química , beta-Arrestinas/metabolismo , Animales , Neoplasias de la Mama/patología , Femenino , Humanos , Transducción de Señal
13.
Methods Enzymol ; 633: 281-294, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32046851

RESUMEN

Measuring protein-protein interactions using purified proteins in vitro is one of the most frequently used approach to understand the biochemical and mechanistic details of cellular signaling pathways. Typically, affinity tags are genetically fused to proteins of interest, and they are used to capture and detect them. However, in some cases, fusion of bulky affinity tags might present a significant limitation in these experiments, especially if the regions in close proximity of tags are involved in protein-protein interactions. Here, we present a step-by-step protocol for an alternative approach that involves reversible biotinylation of purified proteins using a simple chemical-conjugation of cleavable biotin moiety. Biotinylated proteins can be directly used as bait for selective immobilization on solid support for measuring protein-protein interactions. Furthermore, biotinylation of protein of interest also allows specific detection in standard biochemical assays. This simple, straightforward and modular protocol can be directly adapted and applied to facilitate the detection of novel protein-protein interactions as well as measuring apparent affinities of such interactions.


Asunto(s)
Biotina , Proteínas , Biotina/metabolismo , Biotinilación , Proteínas/metabolismo
14.
Methods Enzymol ; 633: 271-280, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32046850

RESUMEN

ß-arrestins (ßarrs) are multifunctional proteins that interact with activated and phosphorylated G protein-coupled receptors (GPCRs) to regulate their signaling and trafficking. Understanding the intricate details of GPCR-ßarr interaction continues to be a key research area in the field of GPCR biology. Bimane fluorescence spectroscopy has been one of the key approaches among a broad range of methods employed to study GPCR-ßarr interaction using purified and reconstituted system. Here, we present a step-by-step protocol for labeling ßarrs with monobromobimane (mBBr) in a site-directed fashion for measuring their interaction with GPCRs and the resulting conformational changes. This simple protocol can be directly applied to other protein-protein interaction modules as well for measuring interactions and conformational changes in reconstituted systems in vitro.


Asunto(s)
Biopelículas , Reactores Biológicos , Compuestos Bicíclicos con Puentes , Receptores Acoplados a Proteínas G , beta-Arrestinas , Proteínas de Unión al GTP , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo
15.
Sci Adv ; 6(37)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32917711

RESUMEN

Agonist-induced phosphorylation of G protein-coupled receptors (GPCRs) is a key determinant for their interaction with ß-arrestins (ßarrs) and subsequent functional responses. Therefore, it is important to decipher the contribution and interplay of different receptor phosphorylation sites in governing ßarr interaction and functional outcomes. Here, we find that several phosphorylation sites in the human vasopressin receptor (V2R), positioned either individually or in clusters, differentially contribute to ßarr recruitment, trafficking, and ERK1/2 activation. Even a single phosphorylation site in V2R, suitably positioned to cross-talk with a key residue in ßarrs, has a decisive contribution in ßarr recruitment, and its mutation results in strong G-protein bias. Molecular dynamics simulation provides mechanistic insights into the pivotal role of this key phosphorylation site in governing the stability of ßarr interaction and regulating the interdomain rotation in ßarrs. Our findings uncover important structural aspects to better understand the framework of GPCR-ßarr interaction and biased signaling.

16.
Structure ; 28(9): 1014-1023.e4, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32579945

RESUMEN

ß-Arrestins (ßarrs) critically regulate G-protein-coupled receptor (GPCR) signaling and trafficking. ßarrs have two isoforms, ßarr1 and ßarr2. Receptor phosphorylation is a key determinant for the binding of ßarrs, and understanding the intricate details of receptor-ßarr interaction is the next frontier in GPCR structural biology. The high-resolution structure of active ßarr1 in complex with a phosphopeptide derived from GPCR has been revealed, but that of ßarr2 remains elusive. Here, we present a 2.3-Å crystal structure of ßarr2 in complex with a phosphopeptide (C7pp) derived from the carboxyl terminus of CXCR7. The structural analysis of C7pp-bound ßarr2 reveals key differences from the previously determined active conformation of ßarr1. One of the key differences is that C7pp-bound ßarr2 shows a relatively small inter-domain rotation. Antibody-fragment-based conformational sensor and hydrogen/deuterium exchange experiments further corroborated the structural features of ßarr2 and suggested that ßarr2 adopts a range of inter-domain rotations.


Asunto(s)
Receptores CXCR/metabolismo , Arrestina beta 2/química , Calorimetría , Cristalografía por Rayos X , Células HEK293 , Humanos , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación , Dominios Proteicos , Transporte de Proteínas/efectos de los fármacos , Receptores CXCR/química , Arrestina beta 2/agonistas , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA