Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.606
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(6): 1527-1546.e25, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38412860

RESUMEN

G protein-coupled receptors (GPCRs) are the largest family of druggable proteins encoded in the human genome, but progress in understanding and targeting them is hindered by the lack of tools to reliably measure their nuanced behavior in physiologically relevant contexts. Here, we developed a collection of compact ONE vector G-protein Optical (ONE-GO) biosensor constructs as a scalable platform that can be conveniently deployed to measure G-protein activation by virtually any GPCR with high fidelity even when expressed endogenously in primary cells. By characterizing dozens of GPCRs across many cell types like primary cardiovascular cells or neurons, we revealed insights into the molecular basis for G-protein coupling selectivity of GPCRs, pharmacogenomic profiles of anti-psychotics on naturally occurring GPCR variants, and G-protein subtype signaling bias by endogenous GPCRs depending on cell type or upon inducing disease-like states. In summary, this open-source platform makes the direct interrogation of context-dependent GPCR activity broadly accessible.


Asunto(s)
Técnicas Biosensibles , Transducción de Señal , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Unión al GTP/metabolismo
2.
Cell ; 187(3): 782-796.e23, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38244547

RESUMEN

The rapid kinetics of biological processes and associated short-lived conformational changes pose a significant challenge in attempts to structurally visualize biomolecules during a reaction in real time. Conventionally, on-pathway intermediates have been trapped using chemical modifications or reduced temperature, giving limited insights. Here, we introduce a time-resolved cryo-EM method using a reusable PDMS-based microfluidic chip assembly with high reactant mixing efficiency. Coating of PDMS walls with SiO2 virtually eliminates non-specific sample adsorption and ensures maintenance of the stoichiometry of the reaction, rendering it highly reproducible. In an operating range from 10 to 1,000 ms, the device allows us to follow in vitro reactions of biological molecules at resolution levels in the range of 3 Å. By employing this method, we show the mechanism of progressive HflX-mediated splitting of the 70S E. coli ribosome in the presence of the GTP via capture of three high-resolution reaction intermediates within 140 ms.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Ribosomas , Microscopía por Crioelectrón/métodos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Unión al GTP/metabolismo , Microfluídica/métodos , Ribosomas/metabolismo , Dióxido de Silicio/análisis
3.
Cell ; 187(6): 1460-1475.e20, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38428423

RESUMEN

Apelin is a key hormone in cardiovascular homeostasis that activates the apelin receptor (APLNR), which is regarded as a promising therapeutic target for cardiovascular disease. However, adverse effects through the ß-arrestin pathway limit its pharmacological use. Here, we report cryoelectron microscopy (cryo-EM) structures of APLNR-Gi1 complexes bound to three agonists with divergent signaling profiles. Combined with functional assays, we have identified "twin hotspots" in APLNR as key determinants for signaling bias, guiding the rational design of two exclusive G-protein-biased agonists WN353 and WN561. Cryo-EM structures of WN353- and WN561-stimulated APLNR-G protein complexes further confirm that the designed ligands adopt the desired poses. Pathophysiological experiments have provided evidence that WN561 demonstrates superior therapeutic effects against cardiac hypertrophy and reduced adverse effects compared with the established APLNR agonists. In summary, our designed APLNR modulator may facilitate the development of next-generation cardiovascular medications.


Asunto(s)
Receptores de Apelina , Fármacos Cardiovasculares , Diseño de Fármacos , Receptores de Apelina/agonistas , Receptores de Apelina/química , Receptores de Apelina/ultraestructura , Microscopía por Crioelectrón , Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Humanos , Fármacos Cardiovasculares/química
4.
Cell ; 186(15): 3208-3226.e27, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37379838

RESUMEN

N7-methylguanosine (m7G) modification, routinely occurring at mRNA 5' cap or within tRNAs/rRNAs, also exists internally in messenger RNAs (mRNAs). Although m7G-cap is essential for pre-mRNA processing and protein synthesis, the exact role of mRNA internal m7G modification remains elusive. Here, we report that mRNA internal m7G is selectively recognized by Quaking proteins (QKIs). By transcriptome-wide profiling/mapping of internal m7G methylome and QKI-binding sites, we identified more than 1,000 high-confidence m7G-modified and QKI-bound mRNA targets with a conserved "GANGAN (N = A/C/U/G)" motif. Strikingly, QKI7 interacts (via C terminus) with the stress granule (SG) core protein G3BP1 and shuttles internal m7G-modified transcripts into SGs to regulate mRNA stability and translation under stress conditions. Specifically, QKI7 attenuates the translation efficiency of essential genes in Hippo signaling pathways to sensitize cancer cells to chemotherapy. Collectively, we characterized QKIs as mRNA internal m7G-binding proteins that modulate target mRNA metabolism and cellular drug resistance.


Asunto(s)
ADN Helicasas , ARN Helicasas , ADN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/genética , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , ARN Helicasas/metabolismo , Gránulos de Estrés , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión al GTP/metabolismo , ARN Mensajero/metabolismo , Gránulos Citoplasmáticos/metabolismo
5.
Cell ; 185(23): 4361-4375.e19, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36368306

RESUMEN

Morphine and fentanyl are among the most used opioid drugs that confer analgesia and unwanted side effects through both G protein and arrestin signaling pathways of µ-opioid receptor (µOR). Here, we report structures of the human µOR-G protein complexes bound to morphine and fentanyl, which uncover key differences in how they bind the receptor. We also report structures of µOR bound to TRV130, PZM21, and SR17018, which reveal preferential interactions of these agonists with TM3 side of the ligand-binding pocket rather than TM6/7 side. In contrast, morphine and fentanyl form dual interactions with both TM3 and TM6/7 regions. Mutations at the TM6/7 interface abolish arrestin recruitment of µOR promoted by morphine and fentanyl. Ligands designed to reduce TM6/7 interactions display preferential G protein signaling. Our results provide crucial insights into fentanyl recognition and signaling of µOR, which may facilitate rational design of next-generation analgesics.


Asunto(s)
Fentanilo , Morfina , Humanos , Analgésicos Opioides/farmacología , Arrestina/metabolismo , Fentanilo/farmacología , Proteínas de Unión al GTP/metabolismo , Morfina/farmacología , Receptores Opioides mu
6.
Cell ; 184(4): 931-942.e18, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571431

RESUMEN

The D1- and D2-dopamine receptors (D1R and D2R), which signal through Gs and Gi, respectively, represent the principal stimulatory and inhibitory dopamine receptors in the central nervous system. D1R and D2R also represent the main therapeutic targets for Parkinson's disease, schizophrenia, and many other neuropsychiatric disorders, and insight into their signaling is essential for understanding both therapeutic and side effects of dopaminergic drugs. Here, we report four cryoelectron microscopy (cryo-EM) structures of D1R-Gs and D2R-Gi signaling complexes with selective and non-selective dopamine agonists, including two currently used anti-Parkinson's disease drugs, apomorphine and bromocriptine. These structures, together with mutagenesis studies, reveal the conserved binding mode of dopamine agonists, the unique pocket topology underlying ligand selectivity, the conformational changes in receptor activation, and potential structural determinants for G protein-coupling selectivity. These results provide both a molecular understanding of dopamine signaling and multiple structural templates for drug design targeting the dopaminergic system.


Asunto(s)
Receptores de Dopamina D1/química , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/química , Receptores de Dopamina D2/metabolismo , Transducción de Señal , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Secuencia de Aminoácidos , Secuencia Conservada , Microscopía por Crioelectrón , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Dopamina D1/ultraestructura , Receptores de Dopamina D2/ultraestructura , Homología Estructural de Proteína
7.
Cell ; 180(6): 1041-1043, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32169216

RESUMEN

ß-arrestins (ßarrs) play multifaceted roles in the signaling and regulation of G-protein-coupled receptors (GPCRs) including their desensitization and endocytosis. Recently determined cryo-EM structures of two different GPCRs in complex with ßarr1 provide the first glimpse of GPCR-ßarr engagement and a structural framework to understand their interaction.


Asunto(s)
Receptores Acoplados a Proteínas G/ultraestructura , beta-Arrestinas/metabolismo , beta-Arrestinas/ultraestructura , Arrestinas/metabolismo , Endocitosis/fisiología , Proteínas de Unión al GTP/metabolismo , Humanos , Fosforilación , Unión Proteica , Isoformas de Proteínas/ultraestructura , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Relación Estructura-Actividad , beta-Arrestina 1/metabolismo , Arrestina beta 2/metabolismo
8.
Cell ; 176(1-2): 306-317.e16, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30503212

RESUMEN

Trypanosome parasites control their virulence and spread by using quorum sensing (QS) to generate transmissible "stumpy forms" in their host bloodstream. However, the QS signal "stumpy induction factor" (SIF) and its reception mechanism are unknown. Although trypanosomes lack G protein-coupled receptor signaling, we have identified a surface GPR89-family protein that regulates stumpy formation. TbGPR89 is expressed on bloodstream "slender form" trypanosomes, which receive the SIF signal, and when ectopically expressed, TbGPR89 drives stumpy formation in a SIF-pathway-dependent process. Structural modeling of TbGPR89 predicts unexpected similarity to oligopeptide transporters (POT), and when expressed in bacteria, TbGPR89 transports oligopeptides. Conversely, expression of an E. coli POT in trypanosomes drives parasite differentiation, and oligopeptides promote stumpy formation in vitro. Furthermore, the expression of secreted trypanosome oligopeptidases generates a paracrine signal that accelerates stumpy formation in vivo. Peptidase-generated oligopeptide QS signals being received through TbGPR89 provides a mechanism for both trypanosome SIF production and reception.


Asunto(s)
Proteínas de Transporte de Membrana/fisiología , Percepción de Quorum/fisiología , Trypanosoma/metabolismo , Diferenciación Celular , Secuencia Conservada/genética , Proteínas de Unión al GTP/metabolismo , Proteínas de Transporte de Membrana/genética , Oligopéptidos/genética , Oligopéptidos/fisiología , Filogenia , Proteínas Protozoarias/metabolismo , Percepción de Quorum/genética , Transducción de Señal , Trypanosoma/fisiología , Trypanosoma brucei brucei/metabolismo , Tripanosomiasis Africana/parasitología , Virulencia/fisiología
9.
Nat Immunol ; 22(3): 312-321, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33510463

RESUMEN

Mitochondrial abnormalities have been noted in lupus, but the causes and consequences remain obscure. Autophagy-related genes ATG5, ATG7 and IRGM have been previously implicated in autoimmune disease. We reasoned that failure to clear defective mitochondria via mitophagy might be a foundational driver in autoimmunity by licensing mitochondrial DNA-dependent induction of type I interferon. Here, we show that mice lacking the GTPase IRGM1 (IRGM homolog) exhibited a type I interferonopathy with autoimmune features. Irgm1 deletion impaired the execution of mitophagy with cell-specific consequences. In fibroblasts, mitochondrial DNA soiling of the cytosol induced cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-dependent type I interferon, whereas in macrophages, lysosomal Toll-like receptor 7 was activated. In vivo, Irgm1-/- tissues exhibited mosaic dependency upon nucleic acid receptors. Whereas salivary and lacrimal gland autoimmune pathology was abolished and lung pathology was attenuated by cGAS and STING deletion, pancreatic pathology remained unchanged. These findings reveal fundamental connections between mitochondrial quality control and tissue-selective autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Fibroblastos/metabolismo , Proteínas de Unión al GTP/metabolismo , Mitocondrias/metabolismo , Mitofagia , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Células Cultivadas , Fibroblastos/inmunología , Fibroblastos/patología , Proteínas de Unión al GTP/deficiencia , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Macrófagos/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/genética , Mitocondrias/inmunología , Mitocondrias/patología , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
10.
Immunity ; 56(3): 562-575.e6, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36842431

RESUMEN

Dietary components and metabolites have a profound impact on immunity and inflammation. Here, we investigated how sensing of cholesterol metabolite oxysterols by γδ T cells impacts their tissue residency and function. We show that dermal IL-17-producing γδ T (Tγδ17) cells essential for skin-barrier homeostasis require oxysterols sensing through G protein receptor 183 (GPR183) for their development and inflammatory responses. Single-cell transcriptomics and murine reporter strains revealed that GPR183 on developing γδ thymocytes is needed for their maturation by sensing medullary thymic epithelial-cell-derived oxysterols. In the skin, basal keratinocytes expressing the oxysterol enzyme cholesterol 25-hydroxylase (CH25H) maintain dermal Tγδ17 cells. Diet-driven increases in oxysterols exacerbate Tγδ17-cell-mediated psoriatic inflammation, dependent on GPR183 on γδ T cells. Hence, cholesterol-derived oxysterols control spatially distinct but biologically linked processes of thymic education and peripheral function of dermal T cells, implicating diet as a focal parameter of dermal Tγδ17 cells.


Asunto(s)
Colesterol en la Dieta , Oxiesteroles , Humanos , Animales , Ratones , Oxiesteroles/metabolismo , Piel/metabolismo , Inflamación , Proteínas de Unión al GTP/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
11.
Cell ; 171(5): 1165-1175.e13, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29149605

RESUMEN

Biased agonism has been proposed as a means to separate desirable and adverse drug responses downstream of G protein-coupled receptor (GPCR) targets. Herein, we describe structural features of a series of mu-opioid-receptor (MOR)-selective agonists that preferentially activate receptors to couple to G proteins or to recruit ßarrestin proteins. By comparing relative bias for MOR-mediated signaling in each pathway, we demonstrate a strong correlation between the respiratory suppression/antinociception therapeutic window in a series of compounds spanning a wide range of signaling bias. We find that ßarrestin-biased compounds, such as fentanyl, are more likely to induce respiratory suppression at weak analgesic doses, while G protein signaling bias broadens the therapeutic window, allowing for antinociception in the absence of respiratory suppression.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Receptores Opioides mu/agonistas , Animales , Fentanilo/administración & dosificación , Proteínas de Unión al GTP/metabolismo , Ratones , Morfina/administración & dosificación , Receptores Opioides mu/química , Sistema Respiratorio/efectos de los fármacos , Transducción de Señal , beta-Arrestinas/metabolismo
13.
Cell ; 167(3): 739-749.e11, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27720449

RESUMEN

G protein-coupled receptor (GPCR) signaling, mediated by hetero-trimeric G proteins, can be differentially controlled by agonists. At a molecular level, this is thought to occur principally via stabilization of distinct receptor conformations by individual ligands. These distinct conformations control subsequent recruitment of transducer and effector proteins. Here, we report that ligand efficacy at the calcitonin GPCR (CTR) is also correlated with ligand-dependent alterations to G protein conformation. We observe ligand-dependent differences in the sensitivity of the G protein ternary complex to disruption by GTP, due to conformational differences in the receptor-bound G protein hetero-trimer. This results in divergent agonist-dependent receptor-residency times for the hetero-trimeric G protein and different accumulation rates for downstream second messengers. This study demonstrates that factors influencing efficacy extend beyond receptor conformation(s) and expands understanding of the molecular basis for how G proteins control/influence efficacy. This has important implications for the mechanisms that underlie ligand-mediated biased agonism. VIDEO ABSTRACT.


Asunto(s)
Proteínas de Unión al GTP/química , Guanosina Trifosfato/farmacología , Receptores de Calcitonina/agonistas , Receptores de Calcitonina/química , Adenosina Difosfato/biosíntesis , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Ligandos , Conformación Proteica , Multimerización de Proteína , Receptores de Calcitonina/metabolismo
14.
Mol Cell ; 83(21): 3852-3868.e6, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37852256

RESUMEN

The Chaperonin Containing Tailless polypeptide 1 (CCT) complex is an essential protein folding machine with a diverse clientele of substrates, including many proteins with ß-propeller domains. Here, we determine the structures of human CCT in complex with its accessory co-chaperone, phosducin-like protein 1 (PhLP1), in the process of folding Gß5, a component of Regulator of G protein Signaling (RGS) complexes. Cryoelectron microscopy (cryo-EM) and image processing reveal an ensemble of distinct snapshots that represent the folding trajectory of Gß5 from an unfolded molten globule to a fully folded ß-propeller. These structures reveal the mechanism by which CCT directs Gß5 folding through initiating specific intermolecular contacts that facilitate the sequential folding of individual ß sheets until the propeller closes into its native structure. This work directly visualizes chaperone-mediated protein folding and establishes that CCT orchestrates folding by stabilizing intermediates through interactions with surface residues that permit the hydrophobic core to coalesce into its folded state.


Asunto(s)
Proteínas de Unión al GTP , Chaperonas Moleculares , Humanos , Microscopía por Crioelectrón , Chaperonas Moleculares/metabolismo , Proteínas de Unión al GTP/metabolismo , Pliegue de Proteína , Transducción de Señal , Chaperoninas
15.
Cell ; 160(6): 1209-21, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25728666

RESUMEN

Rice is sensitive to cold and can be grown only in certain climate zones. Human selection of japonica rice has extended its growth zone to regions with lower temperature, while the molecular basis of this adaptation remains unknown. Here, we identify the quantitative trait locus COLD1 that confers chilling tolerance in japonica rice. Overexpression of COLD1(jap) significantly enhances chilling tolerance, whereas rice lines with deficiency or downregulation of COLD1(jap) are sensitive to cold. COLD1 encodes a regulator of G-protein signaling that localizes on plasma membrane and endoplasmic reticulum (ER). It interacts with the G-protein α subunit to activate the Ca(2+) channel for sensing low temperature and to accelerate G-protein GTPase activity. We further identify that a SNP in COLD1, SNP2, originated from Chinese Oryza rufipogon, is responsible for the ability of COLD(jap/ind) to confer chilling tolerance, supporting the importance of COLD1 in plant adaptation.


Asunto(s)
Proteínas y Péptidos de Choque por Frío/metabolismo , Oryza/fisiología , Proteínas de Plantas/metabolismo , Secuencia de Aminoácidos , Cruzamiento , Proteínas y Péptidos de Choque por Frío/genética , Frío , Retículo Endoplásmico , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica de las Plantas , Datos de Secuencia Molecular , Mutación , Oryza/citología , Oryza/genética , Proteínas de Plantas/química , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Alineación de Secuencia
16.
Mol Cell ; 82(22): 4340-4352.e6, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36309016

RESUMEN

Adhesion G-protein-coupled receptors (aGPCRs) play key roles in a diversity of physiologies. A hallmark of aGPCR activation is the removal of the inhibitory GAIN domain and the dipping of the cleaved stalk peptide into the ligand-binding pocket of receptors; however, the detailed mechanism remains obscure. Here, we present cryoelectron microscopy (cryo-EM) structures of ADGRL3 in complex with Gq, Gs, Gi, and G12. The structures reveal unique ligand-engaging mode, distinctive activation conformation, and key mechanisms of aGPCR activation. The structures also reveal the uncharted structural information of GPCR/G12 coupling. A comparison of Gq, Gs, Gi, and G12 engagements with ADGRL3 reveals the key determinant of G-protein coupling on the far end of αH5 of Gα. A detailed analysis of the engagements allows us to design mutations that specifically enhance one pathway over others. Taken together, our study lays the groundwork for understanding aGPCR activation and G-protein-coupling selectivity.


Asunto(s)
Proteínas de Unión al GTP , Receptores Acoplados a Proteínas G , Ligandos , Microscopía por Crioelectrón , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Unión al GTP/metabolismo
17.
Mol Cell ; 82(14): 2681-2695.e6, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35714614

RESUMEN

Serotonin (or 5-hydroxytryptamine, 5-HT) is an important neurotransmitter that activates 12 different G protein-coupled receptors (GPCRs) through selective coupling of Gs, Gi, or Gq proteins. The structural basis for G protein subtype selectivity by these GPCRs remains elusive. Here, we report the structures of the serotonin receptors 5-HT4, 5-HT6, and 5-HT7 with Gs, and 5-HT4 with Gi1. The structures reveal that transmembrane helices TM5 and TM6 alternate lengths as a macro-switch to determine receptor's selectivity for Gs and Gi, respectively. We find that the macro-switch by the TM5-TM6 length is shared by class A GPCR-G protein structures. Furthermore, we discover specific residues within TM5 and TM6 that function as micro-switches to form specific interactions with Gs or Gi. Together, these results present a common mechanism of Gs versus Gi protein coupling selectivity or promiscuity by class A GPCRs and extend the basis of ligand recognition at serotonin receptors.


Asunto(s)
Receptores Acoplados a Proteínas G , Serotonina , Proteínas de Unión al GTP/metabolismo , Ligandos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo
18.
Cell ; 157(2): 459-471, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725411

RESUMEN

KRas is a major proto-oncogene product whose signaling activity depends on its level of enrichment on the plasma membrane (PM). This PM localization relies on posttranslational prenylation for membrane affinity, while PM specificity has been attributed to electrostatic interactions between negatively charged phospholipids in the PM and basic amino-acids in the C terminus of KRas. By measuring kinetic parameters of KRas dynamics in living cells with a cellular-automata-based data-fitting approach in realistic cell-geometries, we show that charge-based specificity is not sufficient to generate PM enrichment in light of the total surface area of endomembranes. Instead, mislocalized KRas is continuously sequestered from endomembranes by cytosolic PDEδ to be unloaded in an Arl2-dependent manner to perinuclear membranes. Electrostatic interactions then trap KRas at the recycling endosome (RE), from where vesicular transport restores enrichment on the PM. This energy driven reaction-diffusion cycle explains how small molecule targeting of PDEδ affects the spatial organization of KRas.


Asunto(s)
Membrana Celular/metabolismo , Endosomas/metabolismo , Proteínas ras/metabolismo , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Animales , Perros , Proteínas de Unión al GTP/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Células de Riñón Canino Madin Darby , Proto-Oncogenes Mas
19.
Cell ; 159(5): 1086-1095, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416947

RESUMEN

Fighting viral infections is hampered by the scarcity of viral targets and their variability, resulting in development of resistance. Viruses depend on cellular molecules-which are attractive alternative targets-for their life cycle, provided that they are dispensable for normal cell functions. Using the model organism Drosophila melanogaster, we identify the ribosomal protein RACK1 as a cellular factor required for infection by internal ribosome entry site (IRES)-containing viruses. We further show that RACK1 is an essential determinant for hepatitis C virus translation and infection, indicating that its function is conserved for distantly related human and fly viruses. Inhibition of RACK1 does not affect Drosophila or human cell viability and proliferation, and RACK1-silenced adult flies are viable, indicating that this protein is not essential for general translation. Our findings demonstrate a specific function for RACK1 in selective mRNA translation and uncover a target for the development of broad antiviral intervention.


Asunto(s)
Dicistroviridae/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/virología , Proteínas de Unión al GTP/metabolismo , Hepatocitos/virología , Virus de Insectos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Línea Celular Tumoral , Drosophila melanogaster/metabolismo , Hepacivirus/metabolismo , Hepatocitos/metabolismo , Humanos , Modelos Moleculares , Factores de Iniciación de Péptidos/metabolismo , Biosíntesis de Proteínas , Receptores de Cinasa C Activada , Secuencias Reguladoras de Ácido Ribonucleico , Replicación Viral
20.
Nature ; 613(7943): 383-390, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36599982

RESUMEN

Specific, regulated modification of RNAs is important for proper gene expression1,2. tRNAs are rich with various chemical modifications that affect their stability and function3,4. 7-Methylguanosine (m7G) at tRNA position 46 is a conserved modification that modulates steady-state tRNA levels to affect cell growth5,6. The METTL1-WDR4 complex generates m7G46 in humans, and dysregulation of METTL1-WDR4 has been linked to brain malformation and multiple cancers7-22. Here we show how METTL1 and WDR4 cooperate to recognize RNA substrates and catalyse methylation. A crystal structure of METTL1-WDR4 and cryo-electron microscopy structures of METTL1-WDR4-tRNA show that the composite protein surface recognizes the tRNA elbow through shape complementarity. The cryo-electron microscopy structures of METTL1-WDR4-tRNA with S-adenosylmethionine or S-adenosylhomocysteine along with METTL1 crystal structures provide additional insights into the catalytic mechanism by revealing the active site in multiple states. The METTL1 N terminus couples cofactor binding with conformational changes in the tRNA, the catalytic loop and the WDR4 C terminus, acting as the switch to activate m7G methylation. Thus, our structural models explain how post-translational modifications of the METTL1 N terminus can regulate methylation. Together, our work elucidates the core and regulatory mechanisms underlying m7G modification by METTL1, providing the framework to understand its contribution to biology and disease.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Unión al GTP , Metilación , Metiltransferasas , Procesamiento Postranscripcional del ARN , ARN de Transferencia , Humanos , Dominio Catalítico , Cristalografía por Rayos X , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/ultraestructura , Metiltransferasas/química , Metiltransferasas/metabolismo , Metiltransferasas/ultraestructura , ARN de Transferencia/química , ARN de Transferencia/metabolismo , ARN de Transferencia/ultraestructura , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Especificidad por Sustrato , Biocatálisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA