Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4123, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750014

RESUMEN

Avian influenza A viruses (IAVs) pose a public health threat, as they are capable of triggering pandemics by crossing species barriers. Replication of avian IAVs in mammalian cells is hindered by species-specific variation in acidic nuclear phosphoprotein 32 (ANP32) proteins, which are essential for viral RNA genome replication. Adaptive mutations enable the IAV RNA polymerase (FluPolA) to surmount this barrier. Here, we present cryo-electron microscopy structures of monomeric and dimeric avian H5N1 FluPolA with human ANP32B. ANP32B interacts with the PA subunit of FluPolA in the monomeric form, at the site used for its docking onto the C-terminal domain of host RNA polymerase II during viral transcription. ANP32B acts as a chaperone, guiding FluPolA towards a ribonucleoprotein-associated FluPolA to form an asymmetric dimer-the replication platform for the viral genome. These findings offer insights into the molecular mechanisms governing IAV genome replication, while enhancing our understanding of the molecular processes underpinning mammalian adaptations in avian-origin FluPolA.


Asunto(s)
Microscopía por Crioelectrón , Genoma Viral , Subtipo H5N1 del Virus de la Influenza A , Proteínas Nucleares , Replicación Viral , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Replicación Viral/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/química , Animales , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/química , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Proteínas Virales/química , Adaptación Fisiológica/genética , Gripe Humana/virología , ARN Viral/metabolismo , ARN Viral/genética , Células HEK293 , Multimerización de Proteína , Modelos Moleculares
2.
J Virol ; 98(5): e0013824, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38563748

RESUMEN

Influenza A viruses, causing seasonal epidemics and occasional pandemics, rely on interactions with host proteins for their RNA genome transcription and replication. The viral RNA polymerase utilizes host RNA polymerase II (Pol II) and interacts with the serine 5 phosphorylated (pS5) C-terminal domain (CTD) of Pol II to initiate transcription. Our study, using single-particle electron cryomicroscopy (cryo-EM), reveals the structure of the 1918 pandemic influenza A virus polymerase bound to a synthetic pS5 CTD peptide composed of four heptad repeats mimicking the 52 heptad repeat mammalian Pol II CTD. The structure shows that the CTD peptide binds at the C-terminal domain of the PA viral polymerase subunit (PA-C) and reveals a previously unobserved position of the 627 domain of the PB2 subunit near the CTD. We identify crucial residues of the CTD peptide that mediate interactions with positively charged cavities on PA-C, explaining the preference of the viral polymerase for pS5 CTD. Functional analysis of mutants targeting the CTD-binding site within PA-C reveals reduced transcriptional function or defects in replication, highlighting the multifunctional role of PA-C in viral RNA synthesis. Our study provides insights into the structural and functional aspects of the influenza virus polymerase-host Pol II interaction and identifies a target for antiviral development.IMPORTANCEUnderstanding the intricate interactions between influenza A viruses and host proteins is crucial for developing targeted antiviral strategies. This study employs advanced imaging techniques to uncover the structural nuances of the 1918 pandemic influenza A virus polymerase bound to a specific host protein, shedding light on the vital process of viral RNA synthesis. The study identifies key amino acid residues in the influenza polymerase involved in binding host polymerase II (Pol II) and highlights their role in both viral transcription and genome replication. These findings not only deepen our understanding of the influenza virus life cycle but also pinpoint a potential target for antiviral development. By elucidating the structural and functional aspects of the influenza virus polymerase-host Pol II interaction, this research provides a foundation for designing interventions to disrupt viral replication and transcription, offering promising avenues for future antiviral therapies.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Influenza A , ARN Polimerasa II , ARN Polimerasa Dependiente del ARN , Proteínas Virales , Humanos , Virus de la Influenza A/metabolismo , Virus de la Influenza A/genética , Virus de la Influenza A/enzimología , Gripe Humana/virología , Modelos Moleculares , Fosforilación , Unión Proteica , Dominios Proteicos , ARN Polimerasa II/metabolismo , ARN Polimerasa II/química , ARN Viral/metabolismo , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/química , Transcripción Genética , Proteínas Virales/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral
3.
Nat Commun ; 14(1): 7627, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993464

RESUMEN

Human metapneumovirus (HMPV) is a major cause of respiratory illness in young children. The HMPV polymerase (L) binds an obligate cofactor, the phosphoprotein (P). During replication and transcription, the L/P complex traverses the viral RNA genome, which is encapsidated within nucleoproteins (N). An essential interaction between N and a C-terminal region of P tethers the L/P polymerase to the template. This N-P interaction is also involved in the formation of cytoplasmic viral factories in infected cells, called inclusion bodies. To define how the polymerase component P recognizes N-encapsidated RNA (N-RNA) we employed cryogenic electron microscopy (cryo-EM) and molecular dynamics simulations, coupled to activity assays and imaging of inclusion bodies in cells. We report a 2.9 Å resolution structure of a triple-complex between multimeric N, bound to both RNA and the C-terminal region of P. Furthermore, we also present cryo-EM structures of assembled N in different oligomeric states, highlighting the plasticity of N. Combined with our functional assays, these structural data delineate in molecular detail how P attaches to N-RNA whilst retaining substantial conformational dynamics. Moreover, the N-RNA-P triple complex structure provides a molecular blueprint for the design of therapeutics to potentially disrupt the attachment of L/P to its template.


Asunto(s)
Metapneumovirus , Niño , Humanos , Preescolar , Metapneumovirus/genética , Nucleocápside/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Nucleoproteínas/metabolismo , Fosfoproteínas/metabolismo
4.
Nat Commun ; 14(1): 7774, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38012131

RESUMEN

Cryo-electron microscopy (cryo-EM) enables the determination of membrane protein structures in native-like environments. Characterising how membrane proteins interact with the surrounding membrane lipid environment is assisted by resolution of lipid-like densities visible in cryo-EM maps. Nevertheless, establishing the molecular identity of putative lipid and/or detergent densities remains challenging. Here we present LipIDens, a pipeline for molecular dynamics (MD) simulation-assisted interpretation of lipid and lipid-like densities in cryo-EM structures. The pipeline integrates the implementation and analysis of multi-scale MD simulations for identification, ranking and refinement of lipid binding poses which superpose onto cryo-EM map densities. Thus, LipIDens enables direct integration of experimental and computational structural approaches to facilitate the interpretation of lipid-like cryo-EM densities and to reveal the molecular identities of protein-lipid interactions within a bilayer environment. We demonstrate this by application of our open-source LipIDens code to ten diverse membrane protein structures which exhibit lipid-like densities.


Asunto(s)
Proteínas de la Membrana , Simulación de Dinámica Molecular , Proteínas de la Membrana/química , Microscopía por Crioelectrón , Lípidos de la Membrana , Conformación Proteica
5.
Nat Commun ; 14(1): 4160, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37443157

RESUMEN

Infectious protein crystals are an essential part of the viral lifecycle for double-stranded DNA Baculoviridae and double-stranded RNA cypoviruses. These viral protein crystals, termed occlusion bodies or polyhedra, are dense protein assemblies that form a crystalline array, encasing newly formed virions. Here, using X-ray crystallography we determine the structure of a polyhedrin from Nudiviridae. This double-stranded DNA virus family is a sister-group to the baculoviruses, whose members were thought to lack occlusion bodies. The 70-year-old sample contains a well-ordered lattice formed by a predominantly α-helical building block that assembles into a dense, highly interconnected protein crystal. The lattice is maintained by extensive hydrophobic and electrostatic interactions, disulfide bonds, and domain switching. The resulting lattice is resistant to most environmental stresses. Comparison of this structure to baculovirus or cypovirus polyhedra shows a distinct protein structure, crystal space group, and unit cell dimensions, however, all polyhedra utilise common principles of occlusion body assembly.


Asunto(s)
Nudiviridae , Baculoviridae/genética , Proteínas Virales/metabolismo
6.
Sci Adv ; 9(25): eadg7865, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37343087

RESUMEN

Inhibitor discovery for emerging drug-target proteins is challenging, especially when target structure or active molecules are unknown. Here, we experimentally validate the broad utility of a deep generative framework trained at-scale on protein sequences, small molecules, and their mutual interactions-unbiased toward any specific target. We performed a protein sequence-conditioned sampling on the generative foundation model to design small-molecule inhibitors for two dissimilar targets: the spike protein receptor-binding domain (RBD) and the main protease from SARS-CoV-2. Despite using only the target sequence information during the model inference, micromolar-level inhibition was observed in vitro for two candidates out of four synthesized for each target. The most potent spike RBD inhibitor exhibited activity against several variants in live virus neutralization assays. These results establish that a single, broadly deployable generative foundation model for accelerated inhibitor discovery is effective and efficient, even in the absence of target structure or binder information.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Humanos , Anticuerpos Antivirales/química , SARS-CoV-2/metabolismo , Unión Proteica , Secuencia de Aminoácidos
7.
Biomolecules ; 12(7)2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35883427

RESUMEN

Glutamine amidotransferases, enzymes that transfer nitrogen from Gln to various cellular metabolites, are modular, with the amidotransferase (GATase) domain hydrolyzing Gln, generating ammonia and the acceptor domain catalyzing the addition of nitrogen onto its cognate substrate. GMP synthetase (GMPS), an enzyme in the de novo purine nucleotide biosynthetic pathway, is a glutamine amidotransferase that catalyzes the synthesis of GMP from XMP. The reaction involves activation of XMP though adenylation by ATP in the ATP pyrophosphatase (ATPPase) active site, followed by channeling and attack of NH3 generated in the GATase pocket. This complex chemistry entails co-ordination of activity across the active sites, allosteric activation of the GATase domain to modulate Gln hydrolysis and channeling of ammonia from the GATase to the acceptor active site. Functional GMPS dimers associate through the dimerization domain. The crystal structure of the Gln-bound complex of Plasmodium falciparum GMPS (PfGMPS) for the first time revealed large-scale domain rotation to be associated with catalysis and leading to the juxtaposition of two otherwise spatially distal cysteinyl (C113/C337) residues. In this manuscript, we report on an unusual structural variation in the crystal structure of the C89A/C113A PfGMPS double mutant, wherein a larger degree of domain rotation has led to the dissociation of the dimeric structure. Furthermore, we report a hitherto overlooked signature motif tightly related to catalysis.


Asunto(s)
Amoníaco , Ligasas de Carbono-Nitrógeno , Adenosina Trifosfato/química , Amoníaco/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Catálisis , Glutamina/metabolismo , Cinética , Nitrógeno , Conformación Proteica
8.
Nat Commun ; 13(1): 251, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017564

RESUMEN

Influenza A viruses cause seasonal epidemics and global pandemics, representing a considerable burden to healthcare systems. Central to the replication cycle of influenza viruses is the viral RNA-dependent RNA polymerase which transcribes and replicates the viral RNA genome. The polymerase undergoes conformational rearrangements and interacts with viral and host proteins to perform these functions. Here we determine the structure of the 1918 influenza virus polymerase in transcriptase and replicase conformations using cryo-electron microscopy (cryo-EM). We then structurally and functionally characterise the binding of single-domain nanobodies to the polymerase of the 1918 pandemic influenza virus. Combining these functional and structural data we identify five sites on the polymerase which are sensitive to inhibition by nanobodies. We propose that the binding of nanobodies at these sites either prevents the polymerase from assuming particular functional conformations or interactions with viral or host factors. The polymerase is highly conserved across the influenza A subtypes, suggesting these sites as effective targets for potential influenza antiviral development.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/genética , Orthomyxoviridae/genética , Pandemias , Anticuerpos de Dominio Único/química , Animales , Microscopía por Crioelectrón , Genoma Viral , Células HEK293 , Humanos , Virus de la Influenza A/genética , Modelos Moleculares , Unión Proteica , Conformación Proteica , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN , Células Sf9 , Anticuerpos de Dominio Único/genética , Proteínas Virales/química , Proteínas Virales/genética
9.
Mol Cell ; 81(24): 5025-5038.e10, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34890564

RESUMEN

The Sonic Hedgehog (SHH) morphogen pathway is fundamental for embryonic development and stem cell maintenance and is implicated in various cancers. A key step in signaling is transfer of a palmitate group to the SHH N terminus, catalyzed by the multi-pass transmembrane enzyme Hedgehog acyltransferase (HHAT). We present the high-resolution cryo-EM structure of HHAT bound to substrate analog palmityl-coenzyme A and a SHH-mimetic megabody, revealing a heme group bound to HHAT that is essential for HHAT function. A structure of HHAT bound to potent small-molecule inhibitor IMP-1575 revealed conformational changes in the active site that occlude substrate binding. Our multidisciplinary analysis provides a detailed view of the mechanism by which HHAT adapts the membrane environment to transfer an acyl chain across the endoplasmic reticulum membrane. This structure of a membrane-bound O-acyltransferase (MBOAT) superfamily member provides a blueprint for other protein-substrate MBOATs and a template for future drug discovery.


Asunto(s)
Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/metabolismo , Inhibidores Enzimáticos/farmacología , Proteínas Hedgehog/metabolismo , Proteínas de la Membrana/metabolismo , Acilación , Aciltransferasas/genética , Aciltransferasas/ultraestructura , Regulación Alostérica , Animales , Células COS , Dominio Catalítico , Chlorocebus aethiops , Microscopía por Crioelectrón , Células HEK293 , Hemo/metabolismo , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Simulación de Dinámica Molecular , Palmitoil Coenzima A/metabolismo , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad
10.
Glycobiology ; 31(11): 1557-1570, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34245266

RESUMEN

The polysaccharide lyase family 6 (PL6) represents one of the 41 polysaccharide lyase families classified in the CAZy database with the vast majority of its members being alginate lyases grouped into three subfamilies, PL6_1-3. To decipher the mode of recognition and action of the enzymes belonging to subfamily PL6_1, we solved the crystal structures of Pedsa0632, Patl3640, Pedsa3628 and Pedsa3807, which all show different substrate specificities and mode of action (endo-/exolyase). Thorough exploration of the structures of Pedsa0632 and Patl3640 in complex with their substrates as well as docking experiments confirms that the conserved residues in subsites -1 to +3 of the catalytic site form a common platform that can accommodate various types of alginate in a very similar manner but with a series of original adaptations bringing them their specificities of action. From comparative studies with existing structures of PL6_1 alginate lyases, we observe that in the right-handed parallel ß-helix fold shared by all these enzymes, the substrate-binding site harbors the same overall conserved structures and organization. Despite this apparent similarity, it appears that members of the PL6_1 subfamily specifically accommodate and catalyze the degradation of different alginates suggesting that this common platform is actually a highly adaptable and specific tool.


Asunto(s)
Polisacárido Liasas/metabolismo , Secuencia de Aminoácidos , Conformación de Carbohidratos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Polisacárido Liasas/química , Polisacárido Liasas/aislamiento & purificación , Especificidad por Sustrato
12.
Nat Commun ; 12(1): 1238, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33623019

RESUMEN

Flaviviruses such as Dengue (DENV) or Zika virus (ZIKV) assemble into an immature form within the endoplasmatic reticulum (ER), and are then processed by furin protease in the trans-Golgi. To better grasp maturation, we carry out cryo-EM reconstructions of immature Spondweni virus (SPOV), a human flavivirus of the same serogroup as ZIKV. By employing asymmetric localised reconstruction we push the resolution to 3.8 Å, enabling us to refine an atomic model which includes the crucial furin protease recognition site and a conserved Histidine pH-sensor. For direct comparison, we also solve structures of the mature forms of SPONV and DENV to 2.6 Å and 3.1 Å, respectively. We identify an ordered lipid that is present in only the mature forms of ZIKV, SPOV, and DENV and can bind as a consequence of rearranging amphipathic stem-helices of E during maturation. We propose a structural role for the pocket and suggest it stabilizes mature E.


Asunto(s)
Flavivirus/fisiología , Lípidos/química , Glicoproteínas de Membrana/química , Secuencia de Aminoácidos , Flavivirus/ultraestructura , Modelos Moleculares , Estructura Secundaria de Proteína
13.
Nature ; 587(7835): 638-643, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208942

RESUMEN

Aquatic birds represent a vast reservoir from which new pandemic influenza A viruses can emerge1. Influenza viruses contain a negative-sense segmented RNA genome that is transcribed and replicated by the viral heterotrimeric RNA polymerase (FluPol) in the context of viral ribonucleoprotein complexes2,3. RNA polymerases of avian influenza A viruses (FluPolA) replicate viral RNA inefficiently in human cells because of species-specific differences in acidic nuclear phosphoprotein 32 (ANP32), a family of essential host proteins for FluPol activity4. Host-adaptive mutations, particularly a glutamic-acid-to-lysine mutation at amino acid residue 627 (E627K) in the 627 domain of the PB2 subunit, enable avian FluPolA to overcome this restriction and efficiently replicate viral RNA in the presence of human ANP32 proteins. However, the molecular mechanisms of genome replication and the interplay with ANP32 proteins remain largely unknown. Here we report cryo-electron microscopy structures of influenza C virus polymerase (FluPolC) in complex with human and chicken ANP32A. In both structures, two FluPolC molecules form an asymmetric dimer bridged by the N-terminal leucine-rich repeat domain of ANP32A. The C-terminal low-complexity acidic region of ANP32A inserts between the two juxtaposed PB2 627 domains of the asymmetric FluPolA dimer, suggesting a mechanism for how the adaptive PB2(E627K) mutation enables the replication of viral RNA in mammalian hosts. We propose that this complex represents a replication platform for the viral RNA genome, in which one of the FluPol molecules acts as a replicase while the other initiates the assembly of the nascent replication product into a viral ribonucleoprotein complex.


Asunto(s)
Microscopía por Crioelectrón , Gammainfluenzavirus/enzimología , Interacciones Huésped-Patógeno , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Animales , Pollos/virología , Genoma Viral/genética , Células HEK293 , Humanos , Gammainfluenzavirus/genética , Modelos Moleculares , Proteínas Nucleares/ultraestructura , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Multimerización de Proteína , ARN Viral/biosíntesis , ARN Viral/genética , Proteínas de Unión al ARN/ultraestructura , ARN Polimerasa Dependiente del ARN/ultraestructura , Células Sf9
14.
Nat Commun ; 11(1): 6070, 2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230170

RESUMEN

A Correction to this paper has been published: https://doi.org/10.1038/s41467-020-20006-5.

15.
Nat Commun ; 11(1): 5511, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139731

RESUMEN

Parallel molecular evolution and adaptation are important phenomena commonly observed in viruses. Here, we exploit parallel molecular evolution to understand virulence evolution in avian influenza viruses (AIV). Highly-pathogenic AIVs evolve independently from low-pathogenic ancestors via acquisition of polybasic cleavage sites. Why some AIV lineages but not others evolve in this way is unknown. We hypothesise that the parallel emergence of highly-pathogenic AIV may be facilitated by permissive or compensatory mutations occurring across the viral genome. We combine phylogenetic, statistical and structural approaches to discover parallel mutations in AIV genomes associated with the highly-pathogenic phenotype. Parallel mutations were screened using a statistical test of mutation-phenotype association and further evaluated in the contexts of positive selection and protein structure. Our resulting mutational panel may help to reveal new links between virulence evolution and other traits, and raises the possibility of predicting aspects of AIV evolution.


Asunto(s)
Evolución Molecular , Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Gripe Humana/virología , Virulencia/genética , Animales , Secuencia de Bases/genética , Aves/virología , Conjuntos de Datos como Asunto , Genoma Viral/genética , Humanos , Virus de la Influenza A/genética , Gripe Aviar/transmisión , Gripe Humana/transmisión , Mutación , Filogenia , Estabilidad Proteica , Selección Genética , Alineación de Secuencia , Proteínas Virales/genética
18.
Nat Struct Mol Biol ; 27(10): 950-958, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32737466

RESUMEN

The COVID-19 pandemic has had an unprecedented health and economic impact and there are currently no approved therapies. We have isolated an antibody, EY6A, from an individual convalescing from COVID-19 and have shown that it neutralizes SARS-CoV-2 and cross-reacts with SARS-CoV-1. EY6A Fab binds the receptor binding domain (RBD) of the viral spike glycoprotein tightly (KD of 2 nM), and a 2.6-Å-resolution crystal structure of an RBD-EY6A Fab complex identifies the highly conserved epitope, away from the ACE2 receptor binding site. Residues within this footprint are key to stabilizing the pre-fusion spike. Cryo-EM analyses of the pre-fusion spike incubated with EY6A Fab reveal a complex of the intact spike trimer with three Fabs bound and two further multimeric forms comprising the destabilized spike attached to Fab. EY6A binds what is probably a major neutralizing epitope, making it a candidate therapeutic for COVID-19.


Asunto(s)
Anticuerpos Antivirales/química , Betacoronavirus/química , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Adulto , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Betacoronavirus/inmunología , Betacoronavirus/metabolismo , Sitios de Unión , COVID-19 , Chlorocebus aethiops , Reacciones Cruzadas , Microscopía por Crioelectrón , Cristalografía por Rayos X , Epítopos , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Masculino , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Conformación Proteica , Dominios Proteicos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero
19.
Nat Struct Mol Biol ; 27(9): 846-854, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32661423

RESUMEN

The SARS-CoV-2 virus is more transmissible than previous coronaviruses and causes a more serious illness than influenza. The SARS-CoV-2 receptor binding domain (RBD) of the spike protein binds to the human angiotensin-converting enzyme 2 (ACE2) receptor as a prelude to viral entry into the cell. Using a naive llama single-domain antibody library and PCR-based maturation, we have produced two closely related nanobodies, H11-D4 and H11-H4, that bind RBD (KD of 39 and 12 nM, respectively) and block its interaction with ACE2. Single-particle cryo-EM revealed that both nanobodies bind to all three RBDs in the spike trimer. Crystal structures of each nanobody-RBD complex revealed how both nanobodies recognize the same epitope, which partly overlaps with the ACE2 binding surface, explaining the blocking of the RBD-ACE2 interaction. Nanobody-Fc fusions showed neutralizing activity against SARS-CoV-2 (4-6 nM for H11-H4, 18 nM for H11-D4) and additive neutralization with the SARS-CoV-1/2 antibody CR3022.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral , Receptores Virales/metabolismo , Anticuerpos de Dominio Único/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Neutralizantes/ultraestructura , Anticuerpos Antivirales/metabolismo , Anticuerpos Antivirales/ultraestructura , Afinidad de Anticuerpos , Reacciones Antígeno-Anticuerpo/inmunología , Betacoronavirus/metabolismo , Unión Competitiva , COVID-19 , Microscopía por Crioelectrón , Cristalografía por Rayos X , Epítopos/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Modelos Moleculares , Biblioteca de Péptidos , Peptidil-Dipeptidasa A/ultraestructura , Unión Proteica , Conformación Proteica , Receptores Virales/ultraestructura , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , SARS-CoV-2 , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/ultraestructura , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/ultraestructura
20.
Nat Commun ; 11(1): 3228, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32591529

RESUMEN

Plasmodium falciparum (Pf) relies solely on the salvage pathway for its purine nucleotide requirements, making this pathway indispensable to the parasite. Purine nucleotide levels are regulated by anabolic processes and by nucleotidases that hydrolyse these metabolites into nucleosides. Certain apicomplexan parasites, including Pf, have an IMP-specific-nucleotidase 1 (ISN1). Here we show, by comprehensive substrate screening, that PfISN1 catalyzes the dephosphorylation of inosine monophosphate (IMP) and is allosterically activated by ATP. Crystal structures of tetrameric PfISN1 reveal complex rearrangements of domain organization tightly associated with catalysis. Immunofluorescence microscopy and expression of GFP-fused protein indicate cytosolic localization of PfISN1 and expression in asexual and gametocyte stages of the parasite. With earlier evidence on isn1 upregulation in female gametocytes, the structures reported in this study may contribute to initiate the design for possible transmission-blocking agents.


Asunto(s)
5'-Nucleotidasa/química , 5'-Nucleotidasa/metabolismo , Biocatálisis , Plasmodium falciparum/enzimología , Adenosina Trifosfato/metabolismo , Animales , Apoproteínas/metabolismo , Sitios de Unión , Concentración de Iones de Hidrógeno , Cinética , Magnesio/metabolismo , Ratones Endogámicos BALB C , Modelos Moleculares , Proteínas Mutantes/química , Dominios Proteicos , Estructura Secundaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA