Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 691
Filtrer
1.
Sci Adv ; 10(29): eado9413, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39018393

RÉSUMÉ

Chemical ligation of peptides is increasingly used to generate proteins not readily accessible by recombinant approaches. However, a robust method to ligate "difficult" peptides remains to be developed. Here, we report an enhanced native chemical ligation strategy mediated by peptide conjugation in trifluoroacetic acid (TFA). The conjugation between a carboxyl-terminal peptide thiosalicylaldehyde thioester and a 1,3-dithiol-containing peptide in TFA proceeds rapidly to form a thioacetal-linked intermediate, which is readily converted into the desired native amide bond product through simple postligation treatment. The effectiveness and practicality of the method was demonstrated by the successful synthesis of several challenging proteins, including the SARS-CoV-2 transmembrane Envelope (E) protein and nanobodies. Because of the ability of TFA to dissolve virtually all peptides and prevent the formation of unreactive peptide structures, the method is expected to open new opportunities for synthesizing all families of proteins, particularly those with aggregable or colloidal peptide segments.


Sujet(s)
Peptides , Acide trifluoro-acétique , Acide trifluoro-acétique/composition chimique , Peptides/composition chimique , SARS-CoV-2/composition chimique , Anticorps à domaine unique/composition chimique , Humains , COVID-19/virologie
2.
J Phys Chem B ; 128(29): 7033-7042, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39007765

RÉSUMÉ

Experimental evidence has established that SARS-CoV-2 NSP1 acts as a factor that restricts cellular gene expression and impedes mRNA translation within the ribosome's 40S subunit. However, the precise molecular mechanisms underlying this phenomenon have remained elusive. To elucidate this issue, we employed a combination of all-atom steered molecular dynamics and coarse-grained alchemical simulations to explore the binding affinity of mRNA to the 40S ribosome, both in the presence and absence of SARS-CoV-2 NSP1. Our investigations revealed that the binding of SARS-CoV-2 NSP1 to the 40S ribosome leads to a significant enhancement in the binding affinity of mRNA. This observation, which aligns with experimental findings, strongly suggests that SARS-CoV-2 NSP1 has the capability to inhibit mRNA translation. Furthermore, we identified electrostatic interactions between mRNA and the 40S ribosome as the primary driving force behind mRNA translation. Notably, water molecules were found to play a pivotal role in stabilizing the mRNA-40S ribosome complex, underscoring their significance in this process. We successfully pinpointed the specific SARS-CoV-2 NSP1 residues that play a critical role in triggering the translation arrest.


Sujet(s)
Simulation de dynamique moléculaire , Biosynthèse des protéines , ARN messager , Petite sous-unité du ribosome des eucaryotes , SARS-CoV-2 , Protéines virales non structurales , Protéines virales non structurales/métabolisme , Protéines virales non structurales/composition chimique , Protéines virales non structurales/génétique , ARN messager/métabolisme , ARN messager/génétique , ARN messager/composition chimique , Petite sous-unité du ribosome des eucaryotes/métabolisme , Petite sous-unité du ribosome des eucaryotes/composition chimique , SARS-CoV-2/métabolisme , SARS-CoV-2/composition chimique , SARS-CoV-2/génétique , Liaison aux protéines , Humains , Électricité statique
3.
J Phys Chem B ; 128(29): 7141-7147, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39010661

RÉSUMÉ

The binding of the virus to host cells is the first step in viral infection. Human cell angiotensin converting enzyme 2 (ACE2) is the most popular receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), while other receptors have recently been observed in experiments. Neuropilin-1 protein (NRP1) is one of them, but the mechanism of its binding to the wild type (WT) and different variants of the virus remain unclear at the atomic level. In this work, all-atom umbrella sampling simulations were performed to clarify the binding mechanism of NRP1 to the spike protein fragments 679-685 of the WT, Delta, and Omicron BA.1 variants. We found that the Delta variant binds most strongly to NRP1, while the affinity for Omicron BA.1 slightly decreases for NRP1 compared to that of WT, and the van der Waals interaction plays a key role in stabilizing the studied complexes. The change in the protonation state of the His amino acid results in different binding free energies between variants. Consistent with the experiment, decreasing the pH was shown to increase the binding affinity of the virus to NRP1. Our results indicate that Delta and Omicron mutations not only affect fusogenicity but also affect NRP1 binding. In addition, we argue that viral evolution does not further improve NRP1 binding affinity which remains in the µM range but may increase immune evasion.


Sujet(s)
Simulation de dynamique moléculaire , Neuropiline 1 , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Neuropiline 1/métabolisme , Neuropiline 1/composition chimique , Humains , SARS-CoV-2/composition chimique , SARS-CoV-2/métabolisme , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , COVID-19/virologie , COVID-19/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique
4.
Protein Sci ; 33(8): e5109, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38989563

RÉSUMÉ

Understanding how proteins evolve under selective pressure is a longstanding challenge. The immensity of the search space has limited efforts to systematically evaluate the impact of multiple simultaneous mutations, so mutations have typically been assessed individually. However, epistasis, or the way in which mutations interact, prevents accurate prediction of combinatorial mutations based on measurements of individual mutations. Here, we use artificial intelligence to define the entire functional sequence landscape of a protein binding site in silico, and we call this approach Complete Combinatorial Mutational Enumeration (CCME). By leveraging CCME, we are able to construct a comprehensive map of the evolutionary connectivity within this functional sequence landscape. As a proof of concept, we applied CCME to the ACE2 binding site of the SARS-CoV-2 spike protein receptor binding domain. We selected representative variants from across the functional sequence landscape for testing in the laboratory. We identified variants that retained functionality to bind ACE2 despite changing over 40% of evaluated residue positions, and the variants now escape binding and neutralization by monoclonal antibodies. This work represents a crucial initial stride toward achieving precise predictions of pathogen evolution, opening avenues for proactive mitigation.


Sujet(s)
Angiotensin-converting enzyme 2 , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , SARS-CoV-2/génétique , SARS-CoV-2/composition chimique , SARS-CoV-2/métabolisme , Humains , Sites de fixation , COVID-19/virologie , COVID-19/génétique , Liaison aux protéines , Intelligence artificielle
5.
Org Biomol Chem ; 22(29): 5936-5947, 2024 07 24.
Article de Anglais | MEDLINE | ID: mdl-38973558

RÉSUMÉ

DNA aptamers are oligonucleotides that specifically bind to target molecules, similar to how antibodies bind to antigens. We identified an aptamer named MEZ that is highly specific to the receptor-binding domain, RBD, of the SARS-CoV-2 spike protein from the Wuhan-Hu-1 strain. The SELEX procedure was utilized to enrich the initial 31-mer oligonucleotide library with the target aptamer. The aptamer identification was performed using the novel protocol based on nanopore sequencing developed in this study. The MEZ aptamer was chemically synthesized and tested for binding with the SARS-CoV-2 RBD of the spike protein from different strains. The Kd is 6.5 nM for the complex with the RBD from the Wuhan-Hu-1 strain, which is comparable with known aptamers; the advantage is that the MEZ aptamer is smaller than known analogs. The proposed aptamer is highly selective for the RBD protein from the Wuhan-Hu-1 strain and does not form complexes with the RBD from Beta, Delta and Omicron strains. Experimental and theoretical studies together revealed the molecular mechanism of aptamer binding. The aptamer occupies the same binding site as ACE2 when bound to the RBD. The 3'-end of the MEZ aptamer is important for complex formation and is responsible for the discrimination of the RBD protein from a specific strain. The 5'-end is responsible for the formation of a loop in the 3D structure of the aptamer, which is important for proper binding.


Sujet(s)
Aptamères nucléotidiques , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Aptamères nucléotidiques/composition chimique , SARS-CoV-2/composition chimique , SARS-CoV-2/isolement et purification , Humains , Technique SELEX , Liaison aux protéines , COVID-19/virologie , Sites de fixation
6.
ACS Appl Mater Interfaces ; 16(28): 37275-37287, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38959130

RÉSUMÉ

Titanium dioxide (TiO2) shows significant potential as a self-cleaning material to inactivate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and prevent virus transmission. This study provides insights into the impact of UV-A light on the photocatalytic inactivation of adsorbed SARS-CoV-2 virus-like particles (VLPs) on a TiO2 surface at the molecular and atomic levels. X-ray photoelectron spectroscopy, combined with density functional theory calculations, reveals that spike proteins can adsorb on TiO2 predominantly via their amine and amide functional groups in their amino acids blocks. We employ atomic force microscopy and grazing-incidence small-angle X-ray scattering (GISAXS) to investigate the molecular-scale morphological changes during the inactivation of VLPs on TiO2 under light irradiation. Notably, in situ measurements reveal photoinduced morphological changes of VLPs, resulting in increased particle diameters. These results suggest that the denaturation of structural proteins induced by UV irradiation and oxidation of the virus structure through photocatalytic reactions can take place on the TiO2 surface. The in situ GISAXS measurements under an N2 atmosphere reveal that the virus morphology remains intact under UV light. This provides evidence that the presence of both oxygen and UV light is necessary to initiate photocatalytic reactions on the surface and subsequently inactivate the adsorbed viruses. The chemical insights into the virus inactivation process obtained in this study contribute significantly to the development of solid materials for the inactivation of enveloped viruses.


Sujet(s)
SARS-CoV-2 , Titane , Rayons ultraviolets , Titane/composition chimique , Titane/effets des radiations , SARS-CoV-2/effets des radiations , SARS-CoV-2/composition chimique , Inactivation virale/effets des radiations , Inactivation virale/effets des médicaments et des substances chimiques , Humains , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , COVID-19/virologie , COVID-19/prévention et contrôle , Adsorption , Propriétés de surface
7.
J Colloid Interface Sci ; 673: 202-215, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-38875787

RÉSUMÉ

To address the ongoing challenges posed by the SARS-CoV-2 and potentially stronger viruses in the future, the development of effective methods to fabricate patterned graphene (PG) and other precisely functional products has become a new research frontier. Herein, we modeled the "checkerboard" graphene (CG) and stripped graphene (SG) as representatives of PG, and studied their interaction mechanism with the target protein (Mpro) by molecular dynamics simulation. The calculation results on the binding strength and the root mean square deviation values of the active pocket revealed that PG is an effective platform for adsorption, immobilization, and destruction of Mpro. Specifically, CG is found to promote disruption of the active pocket for Mpro, but the presence of "checkerboard" oxidized regions inhibits the adsorption of Mpro. Meanwhile, the SG can effectively confine Mpro within the non-oxidized strips and enhances their binding strength, but doesn't play well on disrupting the active pocket. Our work not only elucidates the biological effects of PGs, but also provides guidance for their targeted and precise utilization in combating the SARS-CoV-2.


Sujet(s)
COVID-19 , Graphite , Simulation de dynamique moléculaire , SARS-CoV-2 , Graphite/composition chimique , Adsorption , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/composition chimique , COVID-19/virologie , Humains , Protéases 3C des coronavirus/composition chimique , Protéases 3C des coronavirus/antagonistes et inhibiteurs , Protéases 3C des coronavirus/métabolisme , Liaison aux protéines
8.
J Mol Model ; 30(7): 203, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38858279

RÉSUMÉ

CONTEXT: The Omicron, Kappa, and Delta variants are different strains of the SARS-CoV-2 virus. Graphene oxide quantum dots (GOQDs) represent a burgeoning class of oxygen-enriched, zero-dimensional materials characterized by their sub-20-nm dimensions. Exhibiting pronounced quantum confinement and edge effects, GOQDs manifest exceptional physical-chemical attributes. This study delves into the potential of graphene oxide quantum dots, elucidating their inherent properties pertinent to the surface structures of SARS-CoV-2, employing an integrated computational approach for the repositioning of inhibitory agents. METHODS: Following rigorous adjustment tests, a spectrum of divergent bonding conformations emerged, with particular emphasis placed on identifying the conformation exhibiting optimal adjustment scores and interactions. The investigation employed molecular docking simulations integrating affinity energy evaluations, electrostatic potential clouds, molecular dynamics encompassing average square root calculations, and the computation of Gibbs-free energy. These values quantify the strength of interaction between GOQDs and SARS-CoV-2 spike protein variants. The receptor structures were optimized using the CHARM-GUI server employing force field AMBERFF14SB. The algorithm embedded in CHARMM offers an efficient interpolation scheme and automatic step size selection, enhancing the efficiency of the optimization process. The 3D structures of the ligands are constructed and optimized with density functional theory (DFT) method based on the most stable conformer of each binder. Autodock Vina Software (ADV) was utilized, where essential parameters were specified. Electrostatic potential maps (MEPs) provide a visual depiction of molecules' charge distributions and related properties. After this, molecular dynamics simulations employing the CHARM36 force field in Gromacs 2022.2 were conducted to investigate GOs' interactions with surface macromolecules of SARS-CoV-2 in an explicit aqueous environment. Furthermore, our investigation suggests that lower values indicate stronger binding. Notably, GO-E consistently showed the most negative values across interactions with different variants, suggesting a higher affinity compared to other GOQDs (GO-A to GO-D).


Sujet(s)
Graphite , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Boîtes quantiques , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Graphite/composition chimique , SARS-CoV-2/composition chimique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Boîtes quantiques/composition chimique , Humains , Liaison aux protéines , Électricité statique , COVID-19/virologie
9.
Viruses ; 16(6)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38932234

RÉSUMÉ

The thermostability of vaccines, particularly enveloped viral vectored vaccines, remains a challenge to their delivery wherever needed. The freeze-drying of viral vectored vaccines is a promising approach but remains challenging due to the water removal process from the outer and inner parts of the virus. In the case of enveloped viruses, freeze-drying induces increased stress on the envelope, which often leads to the inactivation of the virus. In this study, we designed a method to freeze-dry a recombinant vesicular stomatitis virus (VSV) expressing the SARS-CoV-2 spike glycoprotein. Since the envelope of VSV is composed of 50% lipids and 50% protein, the formulation study focused on both the protein and lipid portions of the vector. Formulations were prepared primarily using sucrose, trehalose, and sorbitol as cryoprotectants; mannitol as a lyoprotectant; and histidine as a buffer. Initially, the infectivity of rVSV-SARS-CoV-2 and the cake stability were investigated at different final moisture content levels. High recovery of the infectious viral titer (~0.5 to 1 log loss) was found at 3-6% moisture content, with no deterioration in the freeze-dried cakes. To further minimize infectious viral titer loss, the composition and concentration of the excipients were studied. An increase from 5 to 10% in both the cryoprotectants and lyoprotectant, together with the addition of 0.5% gelatin, resulted in the improved recovery of the infectious virus titer and stable cake formation. Moreover, the secondary drying temperature of the freeze-drying process showed a significant impact on the infectivity of rVSV-SARS-CoV-2. The infectivity of the vector declined drastically when the temperature was raised above 20 °C. Throughout a long-term stability study, formulations containing 10% sugar (sucrose/trehalose), 10% mannitol, 0.5% gelatin, and 10 mM histidine showed satisfactory stability for six months at 2-8 °C. The development of this freeze-drying process and the optimized formulation minimize the need for a costly cold chain distribution system.


Sujet(s)
Vaccins contre la COVID-19 , Cryoprotecteurs , Lyophilisation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Lyophilisation/méthodes , SARS-CoV-2/immunologie , SARS-CoV-2/composition chimique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/composition chimique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/immunologie , Cryoprotecteurs/composition chimique , Cryoprotecteurs/pharmacologie , Tréhalose/composition chimique , COVID-19/prévention et contrôle , COVID-19/virologie , Animaux , Humains , Mannitol/composition chimique , Saccharose/composition chimique , Cellules Vero , Chlorocebus aethiops , Sorbitol/composition chimique , Stabilité de médicament , Histidine/composition chimique , Virus de la stomatite vésiculeuse de type Indiana/génétique , Vaccins synthétiques/composition chimique , Vaccins synthétiques/immunologie
10.
Nucleic Acids Res ; 52(13): 7971-7986, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-38842942

RÉSUMÉ

We present the nuclear magnetic resonance spectroscopy (NMR) solution structure of the 5'-terminal stem loop 5_SL1 (SL1) of the SARS-CoV-2 genome. SL1 contains two A-form helical elements and two regions with non-canonical structure, namely an apical pyrimidine-rich loop and an asymmetric internal loop with one and two nucleotides at the 5'- and 3'-terminal part of the sequence, respectively. The conformational ensemble representing the averaged solution structure of SL1 was validated using NMR residual dipolar coupling (RDC) and small-angle X-ray scattering (SAXS) data. We show that the internal loop is the major binding site for fragments of low molecular weight. This internal loop of SL1 can be stabilized by an A12-C28 interaction that promotes the transient formation of an A+•C base pair. As a consequence, the pKa of the internal loop adenosine A12 is shifted to 5.8, compared to a pKa of 3.63 of free adenosine. Furthermore, applying a recently developed pH-differential mutational profiling (PD-MaP) approach, we not only recapitulated our NMR findings of SL1 but also unveiled multiple sites potentially sensitive to pH across the 5'-UTR of SARS-CoV-2.


Sujet(s)
Conformation d'acide nucléique , ARN viral , SARS-CoV-2 , SARS-CoV-2/génétique , SARS-CoV-2/composition chimique , SARS-CoV-2/métabolisme , ARN viral/composition chimique , ARN viral/génétique , ARN viral/métabolisme , Concentration en ions d'hydrogène , Humains , Diffusion aux petits angles , COVID-19/virologie , COVID-19/génétique , Spectroscopie par résonance magnétique , Diffraction des rayons X , Sites de fixation , Génome viral , Appariement de bases , Régions 5' non traduites , Modèles moléculaires
11.
Biomol Concepts ; 15(1)2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38872399

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) is a novel disease that had devastating effects on human lives and the country's economies worldwide. This disease shows similar parasitic traits, requiring the host's biomolecules for its survival and propagation. Spike glycoproteins severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 spike protein) located on the surface of the COVID-19 virus serve as a potential hotspot for antiviral drug development based on their structure. COVID-19 virus calls into action the chaperonin system that assists the attacker, hence favoring infection. To investigate the interaction that occurs between SARS-CoV-2 spike protein and human molecular chaperons (HSPA8 and sHSP27), a series of steps were carried out which included sequence attainment and analysis, followed by multiple sequence alignment, homology modeling, and protein-protein docking which we performed using Cluspro to predict the interactions between SARS-CoV-2 spike protein and human molecular chaperones of interest. Our findings depicted that SARS-CoV-2 spike protein consists of three distinct chains, chains A, B, and C, which interact forming hydrogen bonds, hydrophobic interactions, and electrostatic interactions with both human HSPA8 and HSP27 with -828.3 and -827.9 kcal/mol as binding energies for human HSPA8 and -1166.7 and -1165.9 kcal/mol for HSP27.


Sujet(s)
COVID-19 , Simulation de docking moléculaire , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Humains , SARS-CoV-2/métabolisme , SARS-CoV-2/composition chimique , COVID-19/virologie , COVID-19/métabolisme , Animaux , Liaison aux protéines , Protéines du choc thermique/composition chimique , Protéines du choc thermique/métabolisme , Petites protéines du choc thermique/métabolisme , Petites protéines du choc thermique/composition chimique , Séquence d'acides aminés
12.
Elife ; 132024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38864493

RÉSUMÉ

Glycosylation of the SARS-CoV-2 spike (S) protein represents a key target for viral evolution because it affects both viral evasion and fitness. Successful variations in the glycan shield are difficult to achieve though, as protein glycosylation is also critical to folding and structural stability. Within this framework, the identification of glycosylation sites that are structurally dispensable can provide insight into the evolutionary mechanisms of the shield and inform immune surveillance. In this work, we show through over 45 µs of cumulative sampling from conventional and enhanced molecular dynamics (MD) simulations, how the structure of the immunodominant S receptor binding domain (RBD) is regulated by N-glycosylation at N343 and how this glycan's structural role changes from WHu-1, alpha (B.1.1.7), and beta (B.1.351), to the delta (B.1.617.2), and omicron (BA.1 and BA.2.86) variants. More specifically, we find that the amphipathic nature of the N-glycan is instrumental to preserve the structural integrity of the RBD hydrophobic core and that loss of glycosylation at N343 triggers a specific and consistent conformational change. We show how this change allosterically regulates the conformation of the receptor binding motif (RBM) in the WHu-1, alpha, and beta RBDs, but not in the delta and omicron variants, due to mutations that reinforce the RBD architecture. In support of these findings, we show that the binding of the RBD to monosialylated ganglioside co-receptors is highly dependent on N343 glycosylation in the WHu-1, but not in the delta RBD, and that affinity changes significantly across VoCs. Ultimately, the molecular and functional insight we provide in this work reinforces our understanding of the role of glycosylation in protein structure and function and it also allows us to identify the structural constraints within which the glycosylation site at N343 can become a hotspot for mutations in the SARS-CoV-2 S glycan shield.


Sujet(s)
Simulation de dynamique moléculaire , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycosylation , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , SARS-CoV-2/métabolisme , SARS-CoV-2/composition chimique , SARS-CoV-2/génétique , Humains , COVID-19/virologie , COVID-19/métabolisme , Polyosides/métabolisme , Polyosides/composition chimique , Domaines protéiques , Sites de fixation , Conformation des protéines , Mutation
13.
Acta Crystallogr D Struct Biol ; 80(Pt 6): 451-463, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38841886

RÉSUMÉ

Fragment-based drug design using X-ray crystallography is a powerful technique to enable the development of new lead compounds, or probe molecules, against biological targets. This study addresses the need to determine fragment binding orientations for low-occupancy fragments with incomplete electron density, an essential step before further development of the molecule. Halogen atoms play multiple roles in drug discovery due to their unique combination of electronegativity, steric effects and hydrophobic properties. Fragments incorporating halogen atoms serve as promising starting points in hit-to-lead development as they often establish halogen bonds with target proteins, potentially enhancing binding affinity and selectivity, as well as counteracting drug resistance. Here, the aim was to unambiguously identify the binding orientations of fragment hits for SARS-CoV-2 nonstructural protein 1 (nsp1) which contain a combination of sulfur and/or chlorine, bromine and iodine substituents. The binding orientations of carefully selected nsp1 analogue hits were focused on by employing their anomalous scattering combined with Pan-Dataset Density Analysis (PanDDA). Anomalous difference Fourier maps derived from the diffraction data collected at both standard and long-wavelength X-rays were compared. The discrepancies observed in the maps of iodine-containing fragments collected at different energies were attributed to site-specific radiation-damage stemming from the strong X-ray absorption of I atoms, which is likely to cause cleavage of the C-I bond. A reliable and effective data-collection strategy to unambiguously determine the binding orientations of low-occupancy fragments containing sulfur and/or halogen atoms while mitigating radiation damage is presented.


Sujet(s)
Halogènes , SARS-CoV-2 , Soufre , Halogènes/composition chimique , Cristallographie aux rayons X/méthodes , Soufre/composition chimique , SARS-CoV-2/composition chimique , Protéines virales non structurales/composition chimique , Humains , Électrons , Modèles moléculaires , Conception de médicament , Liaison aux protéines , Sites de fixation , COVID-19
14.
J Mol Graph Model ; 131: 108813, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38885553

RÉSUMÉ

Despite the waning threat of the COVID-19 pandemic, its detrimental impact on global health persists. Regardless of natural immunity or immunity obtained through vaccination, emerging variants of the virus continue to undergo mutations and propagate globally. The persistent mutations in SARS-CoV-2, along with the subsequent formation of recombinant sub-variants has become a challenge for researchers and health professionals, raising concerns about the efficacy of current vaccines. Gaining a better understanding of the biochemical interactions between the Spike Protein (RBD) of SARS-CoV-2 variants and the human ACE2 receptor can prove to be beneficial in designing and developing antiviral therapeutics that are equally effective against all strains and emerging variants. Our objective in this study was to investigate the interfacial binding pattern of the SARS-CoV-2 RBD-ACE2 complex of the Wild Type (WT), Omicron, and the Omicron recombinant sub-variant XBB.1.16. We aimed to examine the atomic level factors and observe how mutations influence the interaction between the virus and its host using Molecular Dynamics simulation, MM/GBSA energy calculations, and Principal Component Analysis. Our findings reveal a higher degree of structural deviation and flexibility in XBB.1.16 compared to WT and Omicron. PCA indicated a wider cluster and significant flexibility in the movements of XBB.1.16 which can also be observed in free energy landscapes, while the normal mode analysis revealed converging motions within the RBD-ACE2 complexes which can facilitate the interaction between them. A pattern of decreased binding affinity was observed in case of XBB.1.16 when compared to the WT and Omicron. These observed deviations in XBB.1.16 when compared to its parent lineage Omicron, and WT can be attributed to the mutations specific to it. Collectively, these results enhance our understanding of the impact of mutations on the interaction between this strain and the host, taking us one step closer to designing effective antiviral therapeutics against the continually mutating strains.


Sujet(s)
Angiotensin-converting enzyme 2 , Simulation de dynamique moléculaire , Mutation , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , Humains , SARS-CoV-2/génétique , SARS-CoV-2/composition chimique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme , COVID-19/virologie , Sites de fixation , Simulation de docking moléculaire
15.
Acta Crystallogr D Struct Biol ; 80(Pt 7): 535-550, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38935342

RÉSUMÉ

The formation of a vitrified thin film embedded with randomly oriented macromolecules is an essential prerequisite for cryogenic sample electron microscopy. Most commonly, this is achieved using the plunge-freeze method first described nearly 40 years ago. Although this is a robust method, the behaviour of different macromolecules shows great variation upon freezing and often needs to be optimized to obtain an isotropic, high-resolution reconstruction. For a macromolecule in such a film, the probability of encountering the air-water interface in the time between blotting and freezing and adopting preferred orientations is very high. 3D reconstruction using preferentially oriented particles often leads to anisotropic and uninterpretable maps. Currently, there are no general solutions to this prevalent issue, but several approaches largely focusing on sample preparation with the use of additives and novel grid modifications have been attempted. In this study, the effect of physical and chemical factors on the orientations of macromolecules was investigated through an analysis of selected well studied macromolecules, and important parameters that determine the behaviour of proteins on cryo-EM grids were revealed. These insights highlight the nature of the interactions that cause preferred orientations and can be utilized to systematically address orientation bias for any given macromolecule and to provide a framework to design small-molecule additives to enhance sample stability and behaviour.


Sujet(s)
Cryomicroscopie électronique , Humains , Protéines/composition chimique , Cryomicroscopie électronique/méthodes , Modèles moléculaires , Structure tertiaire des protéines , SARS-CoV-2/composition chimique , Tensioactifs/composition chimique , Structure quaternaire des protéines , Escherichia coli/composition chimique , Température
16.
Phys Chem Chem Phys ; 26(25): 17720-17744, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38869513

RÉSUMÉ

In this study, we combined AlphaFold-based approaches for atomistic modeling of multiple protein states and microsecond molecular simulations to accurately characterize conformational ensembles evolution and binding mechanisms of convergent evolution for the SARS-CoV-2 spike Omicron variants BA.1, BA.2, BA.2.75, BA.3, BA.4/BA.5 and BQ.1.1. We employed and validated several different adaptations of the AlphaFold methodology for modeling of conformational ensembles including the introduced randomized full sequence scanning for manipulation of sequence variations to systematically explore conformational dynamics of Omicron spike protein complexes with the ACE2 receptor. Microsecond atomistic molecular dynamics (MD) simulations provide a detailed characterization of the conformational landscapes and thermodynamic stability of the Omicron variant complexes. By integrating the predictions of conformational ensembles from different AlphaFold adaptations and applying statistical confidence metrics we can expand characterization of the conformational ensembles and identify functional protein conformations that determine the equilibrium dynamics for the Omicron spike complexes with the ACE2. Conformational ensembles of the Omicron RBD-ACE2 complexes obtained using AlphaFold-based approaches for modeling protein states and MD simulations are employed for accurate comparative prediction of the binding energetics revealing an excellent agreement with the experimental data. In particular, the results demonstrated that AlphaFold-generated extended conformational ensembles can produce accurate binding energies for the Omicron RBD-ACE2 complexes. The results of this study suggested complementarities and potential synergies between AlphaFold predictions of protein conformational ensembles and MD simulations showing that integrating information from both methods can potentially yield a more adequate characterization of the conformational landscapes for the Omicron RBD-ACE2 complexes. This study provides insights in the interplay between conformational dynamics and binding, showing that evolution of Omicron variants through acquisition of convergent mutational sites may leverage conformational adaptability and dynamic couplings between key binding energy hotspots to optimize ACE2 binding affinity and enable immune evasion.


Sujet(s)
Angiotensin-converting enzyme 2 , Simulation de dynamique moléculaire , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , SARS-CoV-2/composition chimique , SARS-CoV-2/métabolisme , Humains , Thermodynamique , Conformation des protéines , Sites de fixation , Peptidyl-Dipeptidase A/composition chimique , Peptidyl-Dipeptidase A/métabolisme , COVID-19/virologie
17.
Viruses ; 16(6)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38932204

RÉSUMÉ

The Omicron variant and its sub-lineages are the only current circulating SARS-CoV-2 viruses worldwide. In this study, the conformational stability of the isolated Receptor Binding Domain (RBD) of Omicron's spike protein is examined in detail. The parent Omicron lineage has over ten mutations in the ACE2 binding region of the RBD that are specifically associated with its ß hairpin loop domain. It is demonstrated through biophysical molecular computations that the mutations in the ß hairpin loop domain significantly increase the intra-protein interaction energies of intra-loop and loop-RBD interactions. The interaction energy increases include the formation of new hydrogen bonds in the ß hairpin loop domain that help stabilize this critical ACE2 binding region. Our results also agree with recent experiments on the stability of Omicron's core ß barrel domain, outside of its loop domain, and help demonstrate the overall conformational stability of the Omicron RBD. It is further shown here through dynamic simulations that the unbound state of the Omicron RBD remains closely aligned with the bound state configuration, which was not observed for the wild-type RBD. Overall, these studies demonstrate the significantly increased conformational stability of Omicron over its wild-type configuration and raise a number of questions on whether conformational stability could be a positive selection feature of SARS-CoV-2 viral mutational changes.


Sujet(s)
Angiotensin-converting enzyme 2 , Simulation de dynamique moléculaire , Mutation , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , SARS-CoV-2/génétique , SARS-CoV-2/composition chimique , Humains , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , Conformation des protéines , Domaines protéiques , Stabilité protéique , COVID-19/virologie , Sites de fixation
18.
Elife ; 132024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38941236

RÉSUMÉ

Genetic diversity is a hallmark of RNA viruses and the basis for their evolutionary success. Taking advantage of the uniquely large genomic database of SARS-CoV-2, we examine the impact of mutations across the spectrum of viable amino acid sequences on the biophysical phenotypes of the highly expressed and multifunctional nucleocapsid protein. We find variation in the physicochemical parameters of its extended intrinsically disordered regions (IDRs) sufficient to allow local plasticity, but also observe functional constraints that similarly occur in related coronaviruses. In biophysical experiments with several N-protein species carrying mutations associated with major variants, we find that point mutations in the IDRs can have nonlocal impact and modulate thermodynamic stability, secondary structure, protein oligomeric state, particle formation, and liquid-liquid phase separation. In the Omicron variant, distant mutations in different IDRs have compensatory effects in shifting a delicate balance of interactions controlling protein assembly properties, and include the creation of a new protein-protein interaction interface in the N-terminal IDR through the defining P13L mutation. A picture emerges where genetic diversity is accompanied by significant variation in biophysical characteristics of functional N-protein species, in particular in the IDRs.


Like other types of RNA viruses, the genetic material of SARS-CoV-2 (the agent responsible for COVID-19) is formed of an RNA molecule which is prone to accumulating mutations. This gives SARS-CoV-2 the ability to evolve quickly, and often to remain one step ahead of treatments. Understanding how these mutations shape the behavior of RNA viruses is therefore crucial to keep diseases such as COVID-19 under control. The gene that codes for the protein that 'packages' the genetic information inside SARS-CoV-2 is particularly prone to mutations. This nucleocapsid (N) protein participates in many key processes during the life cycle of the virus, including potentially interfering with the immune response. Exactly how the physical properties of the N-Protein are impacted by the mutations in its genetic sequence remains unclear. To investigate this question, Nguyen et al. predicted the various biophysical properties of different regions of the N-protein based on a computer-based analysis of SARS-CoV-2 genetic databases. This allowed them to determine if specific protein regions were positively or negatively charged in different mutants. The analyses showed that some domains exhibited great variability in their charge between protein variants ­ reflecting the fact that the corresponding genetic sequences showed high levels of plasticity. Other regions remained conserved, however, including across related coronaviruses. Nguyen et al. also conducted biochemical experiments on a range of N-proteins obtained from clinically relevant SARS-CoV-2 variants. Their results highlighted the importance of protein segments with no fixed three-dimensional structure. Mutations in the related sequences created high levels of variation in the physical properties of these 'intrinsically disordered' regions, which had wide-ranging consequences. Some of these genetic changes even gave individual N-proteins the ability to interact with each other in a completely new way. These results shed new light on the relationship between genetic mutations and the variable physical properties of RNA virus proteins. Nguyen et al. hope that this knowledge will eventually help to develop more effective treatments for viral infections.


Sujet(s)
Protéines de la nucléocapside des coronavirus , Mutation , SARS-CoV-2 , SARS-CoV-2/génétique , SARS-CoV-2/composition chimique , SARS-CoV-2/métabolisme , Protéines de la nucléocapside des coronavirus/génétique , Protéines de la nucléocapside des coronavirus/composition chimique , Protéines de la nucléocapside des coronavirus/métabolisme , COVID-19/virologie , COVID-19/génétique , Humains , Protéines intrinsèquement désordonnées/composition chimique , Protéines intrinsèquement désordonnées/génétique , Protéines intrinsèquement désordonnées/métabolisme , Phosphoprotéines/composition chimique , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , Protéines nucléocapside/génétique , Protéines nucléocapside/métabolisme , Protéines nucléocapside/composition chimique , Thermodynamique , Stabilité protéique
19.
Chemistry ; 30(42): e202401897, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38785102

RÉSUMÉ

The SARS-CoV-2 genome has been shown to be m6A methylated at several positions in vivo. Strikingly, a DRACH motif, the recognition motif for adenosine methylation, resides in the core of the transcriptional regulatory leader sequence (TRS-L) at position A74, which is highly conserved and essential for viral discontinuous transcription. Methylation at position A74 correlates with viral pathogenicity. Discontinuous transcription produces a set of subgenomic mRNAs that function as templates for translation of all structural and accessory proteins. A74 is base-paired in the short stem-loop structure 5'SL3 that opens during discontinuous transcription to form long-range RNA-RNA interactions with nascent (-)-strand transcripts at complementary TRS-body sequences. A74 can be methylated by the human METTL3/METTL14 complex in vitro. Here, we investigate its impact on the structural stability of 5'SL3 and the long-range TRS-leader:TRS-body duplex formation necessary for synthesis of subgenomic mRNAs of all four viral structural proteins. Methylation uniformly destabilizes 5'SL3 and long-range duplexes and alters their relative equilibrium populations, suggesting that the m6A74 modification acts as a regulator for the abundance of viral structural proteins due to this destabilization.


Sujet(s)
Adénosine , Methyltransferases , ARN messager , ARN viral , SARS-CoV-2 , Transcription génétique , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , SARS-CoV-2/composition chimique , ARN viral/composition chimique , ARN viral/métabolisme , ARN viral/génétique , Methyltransferases/composition chimique , Methyltransferases/métabolisme , ARN messager/composition chimique , ARN messager/génétique , ARN messager/métabolisme , Humains , Méthylation , Adénosine/composition chimique , Adénosine/analogues et dérivés , Conformation d'acide nucléique , Génome viral
20.
ACS Infect Dis ; 10(6): 2032-2046, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38728322

RÉSUMÉ

SARS-CoV-2 spike (S) proteins undergo extensive glycosylation, aiding in proper folding, enhancing stability, and evading host immune surveillance. In this study, we used mass spectrometric analysis to elucidate the N-glycosylation characteristics and disulfide bonding of recombinant spike proteins derived from the SARS-CoV-2 Omicron variant (B.1.1.529) in comparison with the D614G spike variant. Furthermore, we conducted microsecond-long molecular dynamics simulations on spike proteins to resolve how the different N-glycans impact spike conformational sampling in the two variants. Our findings reveal that the Omicron spike protein maintains an overall resemblance to the D614G spike variant in terms of site-specific glycan processing and disulfide bond formation. Nonetheless, alterations in glycans were observed at certain N-glycosylation sites. These changes, in synergy with mutations within the Omicron spike protein, result in increased surface accessibility of the macromolecule, including the ectodomain, receptor-binding domain, and N-terminal domain. Additionally, mutagenesis and pull-down assays reveal the role of glycosylation of a specific sequon (N149); furthermore, the correlation of MD simulation and HDX-MS identified several high-dynamic areas of the spike proteins. These insights contribute to our understanding of the interplay between structure and function, thereby advancing effective vaccination and therapeutic strategies.


Sujet(s)
Simulation de dynamique moléculaire , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Glycosylation , SARS-CoV-2/composition chimique , SARS-CoV-2/génétique , Humains , Polyosides/composition chimique , Polyosides/métabolisme , COVID-19/virologie , Mutation , Conformation des protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE