Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.586
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Biochem ; 93(1): 339-366, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38346274

RESUMEN

The nicotinic acetylcholine receptor has served, since its biochemical identification in the 1970s, as a model of an allosteric ligand-gated ion channel mediating signal transition at the synapse. In recent years, the application of X-ray crystallography and high-resolution cryo-electron microscopy, together with molecular dynamic simulations of nicotinic receptors and homologs, have opened a new era in the understanding of channel gating by the neurotransmitter. They reveal, at atomic resolution, the diversity and flexibility of the multiple ligand-binding sites, including recently discovered allosteric modulatory sites distinct from the neurotransmitter orthosteric site, and the conformational dynamics of the activation process as a molecular switch linking these multiple sites. The model emerging from these studies paves the way for a new pharmacology based, first, upon the occurrence of an original mode of indirect allosteric modulation, distinct from a steric competition for a single and rigid binding site, and second, the design of drugs that specifically interact with privileged conformations of the receptor such as agonists, antagonists, and desensitizers. Research on nicotinic receptors is still at the forefront of understanding the mode of action of drugs on the nervous system.


Asunto(s)
Sitio Alostérico , Microscopía por Crioelectrón , Simulación de Dinámica Molecular , Receptores Nicotínicos , Transducción de Señal , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Regulación Alostérica , Humanos , Animales , Cristalografía por Rayos X , Sitios de Unión , Conformación Proteica , Ligandos , Modelos Moleculares , Multimerización de Proteína , Agonistas Nicotínicos/química , Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/metabolismo
2.
Annu Rev Biochem ; 93(1): 163-187, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38594919

RESUMEN

Positive-strand RNA viruses encompass a variety of established and emerging eukaryotic pathogens. Their genome replication is confined to specialized cytoplasmic membrane compartments known as replication organelles (ROs). These ROs derive from host membranes, transformed into distinct structures such as invaginated spherules or intricate membrane networks including single- and/or double-membrane vesicles. ROs play a vital role in orchestrating viral RNA synthesis and evading detection by innate immune sensors of the host. In recent years, groundbreaking cryo-electron microscopy studies conducted with several prototypic viruses have significantly advanced our understanding of RO structure and function. Notably, these studies unveiled the presence of crown-shaped multimeric viral protein complexes that seem to actively participate in viral RNA synthesis and regulate the release of newly synthesized RNA into the cytosol for translation and packaging. These findings have shed light on novel viral functions and fascinating macromolecular complexes that delineate promising new avenues for future research.


Asunto(s)
Microscopía por Crioelectrón , ARN Viral , Replicación Viral , Microscopía por Crioelectrón/métodos , ARN Viral/metabolismo , ARN Viral/genética , ARN Viral/química , Humanos , Virus ARN Monocatenarios Positivos/metabolismo , Virus ARN Monocatenarios Positivos/genética , Virus ARN Monocatenarios Positivos/química , Virus ARN Monocatenarios Positivos/ultraestructura , Orgánulos/ultraestructura , Orgánulos/virología , Orgánulos/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/ultraestructura , Animales , Compartimentos de Replicación Viral/metabolismo , Compartimentos de Replicación Viral/ultraestructura
3.
Annu Rev Biochem ; 93(1): 189-210, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38768392

RESUMEN

During the last ten years, developments in cryo-electron microscopy have transformed our understanding of eukaryotic ribosome assembly. As a result, the field has advanced from a list of the vast array of ribosome assembly factors toward an emerging molecular movie in which individual frames are represented by structures of stable ribosome assembly intermediates with complementary biochemical and genetic data. In this review, we discuss the mechanisms driving the assembly of yeast and human small and large ribosomal subunits. A particular emphasis is placed on the most recent findings that illustrate key concepts of ribosome assembly, such as folding of preribosomal RNA, the enforced chronology of assembly, enzyme-mediated irreversible transitions, and proofreading of preribosomal particles.


Asunto(s)
Microscopía por Crioelectrón , Proteínas Ribosómicas , Ribosomas , Humanos , Ribosomas/metabolismo , Ribosomas/ultraestructura , Ribosomas/química , Ribosomas/genética , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/química , Proteínas Ribosómicas/genética , ARN Ribosómico/metabolismo , ARN Ribosómico/química , ARN Ribosómico/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Modelos Moleculares , Células Eucariotas/metabolismo , Células Eucariotas/ultraestructura , Pliegue del ARN , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/química , Subunidades Ribosómicas Pequeñas de Eucariotas/genética , Subunidades Ribosómicas Pequeñas de Eucariotas/ultraestructura , Animales
4.
Cell ; 187(20): 5497-5499, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39366340

RESUMEN

In this issue of Cell, Penzes et al. describe the use of cryo-EM to identify the cause of a mysterious disease affecting farmed superworms across the US. The study illustrates the power of ex vivo cryo-EM, which uses amplification-free samples to advance at once diagnostic, DNA packaging mechanism, and preventative measures.


Asunto(s)
Microscopía por Crioelectrón , Microscopía por Crioelectrón/métodos , Animales , Humanos
5.
Cell ; 187(3): 563-584, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38306982

RESUMEN

Biology spans a continuum of length and time scales. Individual experimental methods only glimpse discrete pieces of this spectrum but can be combined to construct a more holistic view. In this Review, we detail the latest advancements in volume electron microscopy (vEM) and cryo-electron tomography (cryo-ET), which together can visualize biological complexity across scales from the organization of cells in large tissues to the molecular details inside native cellular environments. In addition, we discuss emerging methodologies for integrating three-dimensional electron microscopy (3DEM) imaging with multimodal data, including fluorescence microscopy, mass spectrometry, single-particle analysis, and AI-based structure prediction. This multifaceted approach fills gaps in the biological continuum, providing functional context, spatial organization, molecular identity, and native interactions. We conclude with a perspective on incorporating diverse data into computational simulations that further bridge and extend length scales while integrating the dimension of time.


Asunto(s)
Biología , Microscopía Electrónica , Microscopía por Crioelectrón/métodos , Tomografía con Microscopio Electrónico/métodos , Microscopía Fluorescente , Tiempo , Simulación por Computador
6.
Cell ; 187(5): 1296-1311.e26, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38428397

RESUMEN

Most membrane proteins are modified by covalent addition of complex sugars through N- and O-glycosylation. Unlike proteins, glycans do not typically adopt specific secondary structures and remain very mobile, shielding potentially large fractions of protein surface. High glycan conformational freedom hinders complete structural elucidation of glycoproteins. Computer simulations may be used to model glycosylated proteins but require hundreds of thousands of computing hours on supercomputers, thus limiting routine use. Here, we describe GlycoSHIELD, a reductionist method that can be implemented on personal computers to graft realistic ensembles of glycan conformers onto static protein structures in minutes. Using molecular dynamics simulation, small-angle X-ray scattering, cryoelectron microscopy, and mass spectrometry, we show that this open-access toolkit provides enhanced models of glycoprotein structures. Focusing on N-cadherin, human coronavirus spike proteins, and gamma-aminobutyric acid receptors, we show that GlycoSHIELD can shed light on the impact of glycans on the conformation and activity of complex glycoproteins.


Asunto(s)
Glicoproteínas , Simulación de Dinámica Molecular , Humanos , Microscopía por Crioelectrón , Glicoproteínas/química , Glicosilación , Polisacáridos/química
7.
Cell ; 187(5): 1127-1144.e21, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38428393

RESUMEN

Chloroplasts are green plastids in the cytoplasm of eukaryotic algae and plants responsible for photosynthesis. The plastid-encoded RNA polymerase (PEP) plays an essential role during chloroplast biogenesis from proplastids and functions as the predominant RNA polymerase in mature chloroplasts. The PEP-centered transcription apparatus comprises a bacterial-origin PEP core and more than a dozen eukaryotic-origin PEP-associated proteins (PAPs) encoded in the nucleus. Here, we determined the cryo-EM structures of Nicotiana tabacum (tobacco) PEP-PAP apoenzyme and PEP-PAP transcription elongation complexes at near-atomic resolutions. Our data show the PEP core adopts a typical fold as bacterial RNAP. Fifteen PAPs bind at the periphery of the PEP core, facilitate assembling the PEP-PAP supercomplex, protect the complex from oxidation damage, and likely couple gene transcription with RNA processing. Our results report the high-resolution architecture of the chloroplast transcription apparatus and provide the structural basis for the mechanistic and functional study of transcription regulation in chloroplasts.


Asunto(s)
ARN Polimerasas Dirigidas por ADN , Plastidios , Cloroplastos/metabolismo , Microscopía por Crioelectrón , ARN Polimerasas Dirigidas por ADN/genética , Nicotiana/genética , Fotosíntesis , Plastidios/enzimología
8.
Cell ; 187(5): 1145-1159.e21, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38428394

RESUMEN

Chloroplast genes encoding photosynthesis-associated proteins are predominantly transcribed by the plastid-encoded RNA polymerase (PEP). PEP is a multi-subunit complex composed of plastid-encoded subunits similar to bacterial RNA polymerases (RNAPs) stably bound to a set of nuclear-encoded PEP-associated proteins (PAPs). PAPs are essential to PEP activity and chloroplast biogenesis, but their roles are poorly defined. Here, we present cryoelectron microscopy (cryo-EM) structures of native 21-subunit PEP and a PEP transcription elongation complex from white mustard (Sinapis alba). We identify that PAPs encase the core polymerase, forming extensive interactions that likely promote complex assembly and stability. During elongation, PAPs interact with DNA downstream of the transcription bubble and with the nascent mRNA. The models reveal details of the superoxide dismutase, lysine methyltransferase, thioredoxin, and amino acid ligase enzymes that are subunits of PEP. Collectively, these data provide a foundation for the mechanistic understanding of chloroplast transcription and its role in plant growth and adaptation.


Asunto(s)
ARN Polimerasas Dirigidas por ADN , Plastidios , Proteínas de Arabidopsis/metabolismo , Cloroplastos/metabolismo , Microscopía por Crioelectrón , ARN Polimerasas Dirigidas por ADN/química , Regulación de la Expresión Génica de las Plantas , Proteínas de Plantas/química , Plastidios/enzimología , Transcripción Genética
9.
Cell ; 187(5): 1160-1176.e21, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38382524

RESUMEN

The α7 nicotinic acetylcholine receptor is a pentameric ligand-gated ion channel that plays an important role in cholinergic signaling throughout the nervous system. Its unique physiological characteristics and implications in neurological disorders and inflammation make it a promising but challenging therapeutic target. Positive allosteric modulators overcome limitations of traditional α7 agonists, but their potentiation mechanisms remain unclear. Here, we present high-resolution structures of α7-modulator complexes, revealing partially overlapping binding sites but varying conformational states. Structure-guided functional and computational tests suggest that differences in modulator activity arise from the stable rotation of a channel gating residue out of the pore. We extend the study using a time-resolved cryoelectron microscopy (cryo-EM) approach to reveal asymmetric state transitions for this homomeric channel and also find that a modulator with allosteric agonist activity exploits a distinct channel-gating mechanism. These results define mechanisms of α7 allosteric modulation and activation with implications across the pentameric receptor superfamily.


Asunto(s)
Receptor Nicotínico de Acetilcolina alfa 7 , Humanos , Receptor Nicotínico de Acetilcolina alfa 7/química , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/ultraestructura , Sitios de Unión , Microscopía por Crioelectrón , Inflamación/tratamiento farmacológico , Transducción de Señal , Regulación Alostérica
10.
Cell ; 187(16): 4272-4288.e20, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39013469

RESUMEN

Vesicle trafficking is a fundamental process that allows for the sorting and transport of specific proteins (i.e., "cargoes") to different compartments of eukaryotic cells. Cargo recognition primarily occurs through coats and the associated proteins at the donor membrane. However, it remains unclear whether cargoes can also be selected at other stages of vesicle trafficking to further enhance the fidelity of the process. The WDR11-FAM91A1 complex functions downstream of the clathrin-associated AP-1 complex to facilitate protein transport from endosomes to the TGN. Here, we report the cryo-EM structure of human WDR11-FAM91A1 complex. WDR11 directly and specifically recognizes a subset of acidic clusters, which we term super acidic clusters (SACs). WDR11 complex assembly and its binding to SAC-containing proteins are indispensable for the trafficking of SAC-containing proteins and proper neuronal development in zebrafish. Our studies thus uncover that cargo proteins could be recognized in a sequence-specific manner downstream of a protein coat.


Asunto(s)
Microscopía por Crioelectrón , Transporte de Proteínas , Pez Cebra , Humanos , Animales , Endosomas/metabolismo , Células HEK293 , Células HeLa , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Unión Proteica
11.
Cell ; 187(17): 4621-4636.e18, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39067443

RESUMEN

Bidirectional transport in cilia is carried out by polymers of the IFTA and IFTB protein complexes, called anterograde and retrograde intraflagellar transport (IFT) trains. Anterograde trains deliver cargoes from the cell to the cilium tip, then convert into retrograde trains for cargo export. We set out to understand how the IFT complexes can perform these two directly opposing roles before and after conversion. We use cryoelectron tomography and in situ cross-linking mass spectrometry to determine the structure of retrograde IFT trains and compare it with the known structure of anterograde trains. The retrograde train is a 2-fold symmetric polymer organized around a central thread of IFTA complexes. We conclude that anterograde-to-retrograde remodeling involves global rearrangements of the IFTA/B complexes and requires complete disassembly of the anterograde train. Finally, we describe how conformational changes to cargo-binding sites facilitate unidirectional cargo transport in a bidirectional system.


Asunto(s)
Cilios , Microscopía por Crioelectrón , Flagelos , Flagelos/metabolismo , Flagelos/ultraestructura , Cilios/metabolismo , Transporte Biológico , Chlamydomonas reinhardtii/metabolismo , Modelos Moleculares , Transporte de Proteínas
12.
Cell ; 187(20): 5665-5678.e18, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39208797

RESUMEN

In mammalian cells, two phosphatidylserine (PS) synthases drive PS synthesis. Gain-of-function mutations in the Ptdss1 gene lead to heightened PS production, causing Lenz-Majewski syndrome (LMS). Recently, pharmacological inhibition of PSS1 has been shown to suppress tumorigenesis. Here, we report the cryo-EM structures of wild-type human PSS1 (PSS1WT), the LMS-causing Pro269Ser mutant (PSS1P269S), and PSS1WT in complex with its inhibitor DS55980254. PSS1 contains 10 transmembrane helices (TMs), with TMs 4-8 forming a catalytic core in the luminal leaflet. These structures revealed a working mechanism of PSS1 akin to the postulated mechanisms of the membrane-bound O-acyltransferase family. Additionally, we showed that both PS and DS55980254 can allosterically inhibit PSS1 and that inhibition by DS55980254 activates the SREBP pathways, thus enhancing the expression of LDL receptors and increasing cellular LDL uptake. This work uncovers a mechanism of mammalian PS synthesis and suggests that selective PSS1 inhibitors have the potential to lower blood cholesterol levels.


Asunto(s)
Fosfatidilserinas , Humanos , Fosfatidilserinas/metabolismo , Microscopía por Crioelectrón , Lipoproteínas LDL/metabolismo , Receptores de LDL/metabolismo , CDPdiacilglicerol-Serina O-Fosfatidiltransferasa/metabolismo , CDPdiacilglicerol-Serina O-Fosfatidiltransferasa/genética , Animales , Células HEK293
13.
Cell ; 187(17): 4751-4769.e25, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39089252

RESUMEN

The Duffy antigen receptor is a seven-transmembrane (7TM) protein expressed primarily at the surface of red blood cells and displays strikingly promiscuous binding to multiple inflammatory and homeostatic chemokines. It serves as the basis of the Duffy blood group system in humans and also acts as the primary attachment site for malarial parasite Plasmodium vivax and pore-forming toxins secreted by Staphylococcus aureus. Here, we comprehensively profile transducer coupling of this receptor, discover potential non-canonical signaling pathways, and determine the cryoelectron microscopy (cryo-EM) structure in complex with the chemokine CCL7. The structure reveals a distinct binding mode of chemokines, as reflected by relatively superficial binding and a partially formed orthosteric binding pocket. We also observe a dramatic shortening of TM5 and 6 on the intracellular side, which precludes the formation of the docking site for canonical signal transducers, thereby providing a possible explanation for the distinct pharmacological and functional phenotype of this receptor.


Asunto(s)
Microscopía por Crioelectrón , Sistema del Grupo Sanguíneo Duffy , Receptores de Superficie Celular , Humanos , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/química , Sistema del Grupo Sanguíneo Duffy/metabolismo , Sistema del Grupo Sanguíneo Duffy/química , Transducción de Señal , Sitios de Unión , Quimiocinas/metabolismo , Quimiocinas/química , Unión Proteica
14.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38176410

RESUMEN

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Receptores de LDL , Animales , Ratones , Alphavirus/fisiología , Virus de la Encefalitis Equina del Este/fisiología , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/metabolismo , Caballos , Unión Proteica , Receptores de LDL/ultraestructura
15.
Cell ; 187(14): 3638-3651.e18, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38838667

RESUMEN

Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.


Asunto(s)
ADN Polimerasa I , Complejo Shelterina , Proteínas de Unión a Telómeros , Telómero , Humanos , Microscopía por Crioelectrón , ADN Polimerasa I/metabolismo , ADN Primasa/metabolismo , ADN Primasa/genética , Modelos Moleculares , Fosforilación , Complejo Shelterina/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Proteínas de Unión a Telómeros/metabolismo
16.
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
17.
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
18.
Cell ; 187(20): 5572-5586.e15, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39197451

RESUMEN

DNA polymerases are important drug targets, and many structural studies have captured them in distinct conformations. However, a detailed understanding of the impact of polymerase conformational dynamics on drug resistance is lacking. We determined cryoelectron microscopy (cryo-EM) structures of DNA-bound herpes simplex virus polymerase holoenzyme in multiple conformations and interacting with antivirals in clinical use. These structures reveal how the catalytic subunit Pol and the processivity factor UL42 bind DNA to promote processive DNA synthesis. Unexpectedly, in the absence of an incoming nucleotide, we observed Pol in multiple conformations with the closed state sampled by the fingers domain. Drug-bound structures reveal how antivirals may selectively bind enzymes that more readily adopt the closed conformation. Molecular dynamics simulations and the cryo-EM structure of a drug-resistant mutant indicate that some resistance mutations modulate conformational dynamics rather than directly impacting drug binding, thus clarifying mechanisms that drive drug selectivity.


Asunto(s)
Antivirales , Microscopía por Crioelectrón , ADN Polimerasa Dirigida por ADN , Farmacorresistencia Viral , Simulación de Dinámica Molecular , Proteínas Virales , Antivirales/farmacología , Antivirales/química , Antivirales/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/química , Proteínas Virales/metabolismo , Proteínas Virales/química , Humanos , ADN Viral/metabolismo , Exodesoxirribonucleasas
19.
Cell ; 187(18): 4996-5009.e14, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38996527

RESUMEN

Latent membrane protein 1 (LMP1) is the primary oncoprotein of Epstein-Barr virus (EBV) and plays versatile roles in the EBV life cycle and pathogenesis. Despite decades of extensive research, the molecular basis for LMP1 folding, assembly, and activation remains unclear. Here, we report cryo-electron microscopy structures of LMP1 in two unexpected assemblies: a symmetric homodimer and a higher-order filamentous oligomer. LMP1 adopts a non-canonical and unpredicted fold that supports the formation of a stable homodimer through tight and antiparallel intermolecular packing. LMP1 dimers further assemble side-by-side into higher-order filamentous oligomers, thereby allowing the accumulation and specific organization of the flexible cytoplasmic tails for efficient recruitment of downstream factors. Super-resolution microscopy and cellular functional assays demonstrate that mutations at both dimeric and oligomeric interfaces disrupt LMP1 higher-order assembly and block multiple LMP1-mediated signaling pathways. Our research provides a framework for understanding the mechanism of LMP1 and for developing potential therapies targeting EBV-associated diseases.


Asunto(s)
Herpesvirus Humano 4 , Proteínas de la Matriz Viral , Humanos , Microscopía por Crioelectrón , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Virus de Epstein-Barr/metabolismo , Células HEK293 , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Modelos Moleculares , Mutación , Multimerización de Proteína , Transducción de Señal , Proteínas de la Matriz Viral/metabolismo , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética
20.
Cell ; 187(16): 4231-4245.e13, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38964328

RESUMEN

The human coronavirus HKU1 spike (S) glycoprotein engages host cell surface sialoglycans and transmembrane protease serine 2 (TMPRSS2) to initiate infection. The molecular basis of HKU1 binding to TMPRSS2 and determinants of host receptor tropism remain elusive. We designed an active human TMPRSS2 construct enabling high-yield recombinant production in human cells of this key therapeutic target. We determined a cryo-electron microscopy structure of the HKU1 RBD bound to human TMPRSS2, providing a blueprint of the interactions supporting viral entry and explaining the specificity for TMPRSS2 among orthologous proteases. We identified TMPRSS2 orthologs from five mammalian orders promoting HKU1 S-mediated entry into cells along with key residues governing host receptor usage. Our data show that the TMPRSS2 binding motif is a site of vulnerability to neutralizing antibodies and suggest that HKU1 uses S conformational masking and glycan shielding to balance immune evasion and receptor engagement.


Asunto(s)
Microscopía por Crioelectrón , Serina Endopeptidasas , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus , Humanos , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Animales , Células HEK293 , Unión Proteica , Anticuerpos Neutralizantes/inmunología , Modelos Moleculares , Receptores Virales/metabolismo , Receptores Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA