Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.657
1.
Bioprocess Biosyst Eng ; 47(6): 877-890, 2024 Jun.
Article En | MEDLINE | ID: mdl-38703202

Ultracentrifugation is an attractive method for separating full and empty capsids, exploiting their density difference. Changes of the serotype/capsid, density of loading material, or the genetic information contained in the adeno-associated viruses (AAVs) require the adaptation of the harvesting parameters and the density gradient loaded onto the centrifuge. To streamline these adaptations, a mathematical model could support the design and testing of operating conditions.Here, hybrid models, which combine empirical functions with artificial neural networks, are proposed to describe the separation of full and empty capsids as a function of material and operational parameters, i.e., the harvest model. In addition, critical quality attributes are estimated by a quality model which is operating on top of the harvest model. The performance of these models was evaluated using test data and two additional blind runs. Also, a "what-if" analysis was conducted to investigate whether the models' predictions align with expectations.It is concluded that the models are sufficiently accurate to support the design of operating conditions, though the accuracy and applicability of the models can further be increased by training them on more specific data with higher variability.


Dependovirus , Ultracentrifugation , Dependovirus/genetics , Dependovirus/isolation & purification , Ultracentrifugation/methods , Virion/isolation & purification , Virion/chemistry , Neural Networks, Computer
2.
Biosens Bioelectron ; 257: 116171, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38636317

The COVID-19 pandemic has highlighted the need for rapid and sensitive detection of SARS-CoV-2. Here, we report an ultrasensitive SARS-CoV-2 immunosensor by integration of an AlGaN/GaN high-electron-mobility transistor (HEMT) and anti-SARS-CoV-2 spike protein antibody. The AlGaN/GaN HEMT immunosensor has demonstrated the capability to detect SARS-CoV-2 spike proteins at an impressively low concentration of 10-22 M. The sensor was also applied to pseudoviruses and SARS-CoV-2 ΔN virions that display the Spike proteins with a single virion particle sensitivity. These features validate the potential of AlGaN/GaN HEMT biosensors for point of care tests targeting SARS-CoV-2. This research not only provides the first HEMT biosensing platform for ultrasensitive and label-free detection of SARS-CoV-2.


Biosensing Techniques , COVID-19 , Gallium , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Transistors, Electronic , Virion , SARS-CoV-2/isolation & purification , SARS-CoV-2/immunology , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/analysis , Humans , COVID-19/diagnosis , COVID-19/virology , Gallium/chemistry , Virion/isolation & purification , Virion/chemistry , Limit of Detection , Aluminum Compounds/chemistry , Equipment Design , Immunoassay/instrumentation , Immunoassay/methods , Antibodies, Immobilized/chemistry , Antibodies, Viral
3.
ACS Sens ; 9(4): 1978-1991, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38564767

This paper presents a sponge-based electrochemical sensor for rapid, on-site collection and analysis of infectious viruses on solid surfaces. The device utilizes a conducting porous sponge modified with graphene, graphene oxide, and specific antibodies. The sponge serves as a hydrophilic porous electrode capable of liquid collection and electrochemical measurements. The device operation involves spraying an aqueous solution on a target surface, swiping the misted surface using the sponge, discharging an electrolyte solution with a simple finger press, and performing in situ incubation and electrochemical measurements. By leveraging the water-absorbing ability of the biofunctionalized conducting sponge, the sensor can effectively collect and quantify virus particles from the surface. The portability of the device is enhanced by introducing a push-release feature that dispenses the liquid electrolyte from a miniature reservoir onto the sensor surface. This reservoir has sharp edges to rupture a liquid sealing film with a finger press. The ability of the device to sample and quantify viral particles is demonstrated by using influenza A virus as the model. The sensor provided a calculated limit of detection of 0.4 TCID50/mL for H1N1 virus, along with a practical concentration range from 1-106 TCID50/mL. Additionally, it achieves a 15% collection efficiency from single-run swiping on a tabletop surface. This versatile device allows for convenient on-site virus detection within minutes, eliminating the need for sample pretreatment and simplifying the entire sample collecting and measuring process. This device presents significant potential for rapid virus detection on solid surfaces.


Electrochemical Techniques , Graphite , Influenza A Virus, H1N1 Subtype , Virion , Graphite/chemistry , Virion/chemistry , Virion/isolation & purification , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Influenza A Virus, H1N1 Subtype/isolation & purification , Surface Properties , Porosity , Electrodes , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Limit of Detection , Humans
4.
J Virol ; 98(4): e0197223, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38470155

The coordinated packaging of the segmented genome of the influenza A virus (IAV) into virions is an essential step of the viral life cycle. This process is controlled by the interaction of packaging signals present in all eight viral RNA (vRNA) segments and the viral nucleoprotein (NP), which binds vRNA via a positively charged binding groove. However, mechanistic models of how the packaging signals and NP work together to coordinate genome packaging are missing. Here, we studied genome packaging in influenza A/SC35M virus mutants that carry mutated packaging signals as well as specific amino acid substitutions at the highly conserved lysine (K) residues 184 and 229 in the RNA-binding groove of NP. Because these lysines are acetylated and thus neutrally charged in infected host cells, we replaced them with glutamine to mimic the acetylated, neutrally charged state or arginine to mimic the non-acetylated, positively charged state. Our analysis shows that the coordinated packaging of eight vRNAs is influenced by (i) the charge state of the replacing amino acid and (ii) its location within the RNA-binding groove. Accordingly, we propose that lysine acetylation induces different charge states within the RNA-binding groove of NP, thereby supporting the activity of specific packaging signals during coordinated genome packaging. IMPORTANCE: Influenza A viruses (IAVs) have a segmented viral RNA (vRNA) genome encapsidated by multiple copies of the viral nucleoprotein (NP) and organized into eight distinct viral ribonucleoprotein complexes. Although genome segmentation contributes significantly to viral evolution and adaptation, it requires a highly sophisticated genome-packaging mechanism. How eight distinct genome complexes are incorporated into the virion is poorly understood, but previous research suggests an essential role for both vRNA packaging signals and highly conserved NP amino acids. By demonstrating that the packaging process is controlled by charge-dependent interactions of highly conserved lysine residues in NP and vRNA packaging signals, our study provides new insights into the sophisticated packaging mechanism of IAVs.


Influenza A virus , Nucleocapsid Proteins , Viral Genome Packaging , Animals , Dogs , Humans , Amino Acid Substitution , Cell Line , Genome, Viral , Influenza A virus/chemistry , Influenza A virus/genetics , Influenza A virus/metabolism , Lysine/genetics , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , RNA, Viral/metabolism , Viral Genome Packaging/genetics , Virion/chemistry , Virion/genetics , Virion/metabolism , Mutation , Static Electricity
5.
Curr Opin Struct Biol ; 86: 102787, 2024 Jun.
Article En | MEDLINE | ID: mdl-38458088

X-ray crystallography and cryo-electron microscopy have enabled the determination of structures of numerous viruses at high resolution and have greatly advanced the field of structural virology. These structures represent only a subset of snapshot end-state conformations, without describing all conformational transitions that virus particles undergo. Allostery plays a critical role in relaying the effects of varied perturbations both on the surface through environmental changes and protein (receptor/antibody) interactions into the genomic core of the virus. Correspondingly, allostery carries implications for communicating changes in genome packaging to the overall stability of the virus particle. Amide hydrogen/deuterium exchange mass spectrometry (HDXMS) of whole viruses is a powerful probe for uncovering virus allostery. Here we critically discuss advancements in understanding virus dynamics by HDXMS with single particle cryo-EM and computational approaches.


Hydrogen Deuterium Exchange-Mass Spectrometry , Virion , Hydrogen Deuterium Exchange-Mass Spectrometry/methods , Virion/chemistry , Virion/metabolism , Allosteric Regulation , Amides/chemistry , Viruses/chemistry , Viruses/metabolism , Cryoelectron Microscopy/methods , Mass Spectrometry/methods , Deuterium Exchange Measurement
6.
Nature ; 627(8005): 905-914, 2024 Mar.
Article En | MEDLINE | ID: mdl-38448589

A string of nucleotides confined within a protein capsid contains all the instructions necessary to make a functional virus particle, a virion. Although the structure of the protein capsid is known for many virus species1,2, the three-dimensional organization of viral genomes has mostly eluded experimental probes3,4. Here we report all-atom structural models of an HK97 virion5, including its entire 39,732 base pair genome, obtained through multiresolution simulations. Mimicking the action of a packaging motor6, the genome was gradually loaded into the capsid. The structure of the packaged capsid was then refined through simulations of increasing resolution, which produced a 26 million atom model of the complete virion, including water and ions confined within the capsid. DNA packaging occurs through a loop extrusion mechanism7 that produces globally different configurations of the packaged genome and gives each viral particle individual traits. Multiple microsecond-long all-atom simulations characterized the effect of the packaged genome on capsid structure, internal pressure, electrostatics and diffusion of water, ions and DNA, and revealed the structural imprints of the capsid onto the genome. Our approach can be generalized to obtain complete all-atom structural models of other virus species, thereby potentially revealing new drug targets at the genome-capsid interface.


Bacteriophages , Capsid , DNA, Viral , Genome, Viral , Virion , Virus Assembly , Bacteriophages/chemistry , Bacteriophages/genetics , Bacteriophages/growth & development , Bacteriophages/metabolism , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Diffusion , DNA, Viral/chemistry , DNA, Viral/genetics , DNA, Viral/metabolism , Ions/analysis , Ions/chemistry , Ions/metabolism , Static Electricity , Virion/chemistry , Virion/genetics , Virion/metabolism , Virus Assembly/genetics , Water/analysis , Water/chemistry , Water/metabolism
7.
J Virol ; 98(3): e0182723, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38305183

Most icosahedral DNA viruses package and condense their genomes into pre-formed, volumetrically constrained capsids. However, concurrent genome biosynthesis and packaging are specific to single-stranded (ss) DNA micro- and parvoviruses. Before packaging, ~120 copies of the øX174 DNA-binding protein J interact with double-stranded DNA. 60 J proteins enter the procapsid with the ssDNA genome, guiding it between 60 icosahedrally ordered DNA-binding pockets formed by the capsid proteins. Although J proteins are small, 28-37 residues in length, they have two domains. The basic, positively charged N-terminus guides the genome between binding pockets, whereas the C-terminus acts as an anchor to the capsid's inner surface. Three C-terminal aromatic residues, W30, Y31, and F37, interact most extensively with the coat protein. Their corresponding codons were mutated, and the resulting strains were biochemically and genetically characterized. Depending on the mutation, the substitutions produced unstable packaging complexes, unstable virions, infectious progeny, or particles packaged with smaller genomes, the latter being a novel phenomenon. The smaller genomes contained internal deletions. The juncture sequences suggest that the unessential A* (A star) protein mediates deletion formation.IMPORTANCEUnessential but strongly conserved gene products are understudied, especially when mutations do not confer discernable phenotypes or the protein's contribution to fitness is too small to reliably determine in laboratory-based assays. Consequently, their functions and evolutionary impact remain obscure. The data presented herein suggest that microvirus A* proteins, discovered over 40 years ago, may hasten the termination of non-productive packaging events. Thus, performing a salvage function by liberating the reusable components of the failed packaging complexes, such as DNA templates and replication enzymes.


Bacteriophage phi X 174 , Capsid Proteins , DNA, Single-Stranded , DNA, Viral , DNA-Binding Proteins , Evolution, Molecular , Viral Genome Packaging , Bacteriophage phi X 174/chemistry , Bacteriophage phi X 174/genetics , Bacteriophage phi X 174/growth & development , Bacteriophage phi X 174/metabolism , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Conserved Sequence , DNA, Single-Stranded/metabolism , DNA, Viral/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Fitness , Mutation , Phenotype , Templates, Genetic , Virion/chemistry , Virion/genetics , Virion/growth & development , Virion/metabolism
8.
J Virol ; 98(3): e0153623, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38315014

African swine fever (ASF) is a highly contagious viral disease that affects domestic and wild pigs. The causative agent of ASF is African swine fever virus (ASFV), a large double-stranded DNA virus with a complex virion structure. Among the various proteins encoded by ASFV, A137R is a crucial structural protein associated with its virulence. However, the structure and molecular mechanisms underlying the functions of A137R remain largely unknown. In this study, we present the structure of A137R determined by cryogenic electron microscopy single-particle reconstruction, which reveals that A137R self-oligomerizes to form a dodecahedron-shaped cage composed of 60 polymers. The dodecahedron is literally equivalent to a T = 1 icosahedron where the icosahedral vertexes are located in the center of each dodecahedral facet. Within each facet, five A137R protomers are arranged in a head-to-tail orientation with a long N-terminal helix forming the edge through which adjacent facets stitch together to form the dodecahedral cage. Combining structural analysis and biochemical evidence, we demonstrate that the N-terminal domain of A137R is crucial and sufficient for mediating the assembly of the dodecahedron. These findings imply the role of A137R cage as a core component in the icosahedral ASFV virion and suggest a promising molecular scaffold for nanotechnology applications. IMPORTANCE: African swine fever (ASF) is a lethal viral disease of pigs caused by African swine fever virus (ASFV). No commercial vaccines and antiviral treatments are available for the prevention and control of the disease. A137R is a structural protein of ASFV that is associated with its virulence. The discovery of the dodecahedron-shaped cage structure of A137R in this study is of great importance in understanding ASFV pathogenicity. This finding sheds light on the molecular mechanisms underlying the functions of A137R. Furthermore, the dodecahedral cage formed by A137R shows promise as a molecular scaffold for nanoparticle vectors. Overall, this study provides valuable insights into the structure and function of A137R, contributing to our understanding of ASFV and potentially opening up new avenues for the development of vaccines or treatments for ASF.


African Swine Fever Virus , Swine , Viral Structural Proteins , Animals , African Swine Fever/virology , African Swine Fever Virus/chemistry , African Swine Fever Virus/growth & development , African Swine Fever Virus/pathogenicity , African Swine Fever Virus/ultrastructure , Cryoelectron Microscopy , Structure-Activity Relationship , Swine/virology , Viral Structural Proteins/chemistry , Viral Structural Proteins/metabolism , Viral Structural Proteins/ultrastructure , Virion/chemistry , Virion/metabolism , Virion/ultrastructure , Virulence
9.
J Virol ; 98(3): e0182023, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38329331

Multi-segmented viruses often multimerize their genomic segments to ensure efficient and stoichiometric packaging of the correct genetic cargo. In the bipartite Nodaviridae family, genome heterodimerization is also observed and conserved among different species. However, the nucleotide composition and biological function for this heterodimer remain unclear. Using Flock House virus as a model system, we developed a next-generation sequencing approach ("XL-ClickSeq") to probe heterodimer site sequences. We identified an intermolecular base-pairing site which contributed to heterodimerization in both wild-type and defective virus particles. Mutagenic disruption of this heterodimer site exhibited significant deficiencies in genome packaging and encapsidation specificity to viral genomic RNAs. Furthermore, the disruption of this intermolecular interaction directly impacts the thermostability of the mature virions. These results demonstrate that the intermolecular RNA-RNA interactions within the encapsidated genome of an RNA virus have an important role on virus particle integrity and thus may impact its transmission to a new host.IMPORTANCEFlock House virus is a member of Nodaviridae family of viruses, which provides a well-studied model virus for non-enveloped RNA virus assembly, cell entry, and replication. The Flock House virus genome consists of two separate RNA molecules, which can form a heterodimer upon heating of virus particles. Although similar RNA dimerization is utilized by other viruses (such as retroviruses) as a packaging mechanism and is conserved among Nodaviruses, the role of heterodimerization in the Nodavirus replication cycle is unclear. In this research, we identified the RNA sequences contributing to Flock House virus genome heterodimerization and discovered that such RNA-RNA interaction plays an essential role in virus packaging efficiency and particle integrity. This provides significant insight into how the interaction of packaged viral RNA may have a broader impact on the structural and functional properties of virus particles.


Dimerization , Genome, Viral , Nodaviridae , RNA, Viral , Thermodynamics , Viral Genome Packaging , Virion , Animals , Base Pairing/genetics , Genome, Viral/genetics , High-Throughput Nucleotide Sequencing , Mutation , Nodaviridae/chemistry , Nodaviridae/genetics , Nodaviridae/growth & development , RNA Virus Infections/transmission , RNA Virus Infections/veterinary , RNA Virus Infections/virology , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Viral Genome Packaging/genetics , Virion/chemistry , Virion/genetics , Virion/metabolism
10.
Sci Adv ; 10(8): eadj1640, 2024 Feb 23.
Article En | MEDLINE | ID: mdl-38394211

The compartmentalization of eukaryotic cells presents considerable challenges to the herpesvirus life cycle. The herpesvirus tegument, a bulky proteinaceous aggregate sandwiched between herpesviruses' capsid and envelope, is uniquely evolved to address these challenges, yet tegument structure and organization remain poorly characterized. We use deep-learning-enhanced cryogenic electron microscopy to investigate the tegument of human cytomegalovirus virions and noninfectious enveloped particles (NIEPs; a genome packaging-aborted state), revealing a portal-biased tegumentation scheme. We resolve atomic structures of portal vertex-associated tegument (PVAT) and identify multiple configurations of PVAT arising from layered reorganization of pUL77, pUL48 (large tegument protein), and pUL47 (inner tegument protein) assemblies. Analyses show that pUL77 seals the last-packaged viral genome end through electrostatic interactions, pUL77 and pUL48 harbor a head-linker-capsid-binding motif conducive to PVAT reconfiguration, and pUL47/48 dimers form 45-nm-long filaments extending from the portal vertex. These results provide a structural framework for understanding how herpesvirus tegument facilitates and evolves during processes spanning viral genome packaging to delivery.


Capsid Proteins , Cytomegalovirus , Humans , Cytomegalovirus/chemistry , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cryoelectron Microscopy , Capsid Proteins/chemistry , Capsid/chemistry , Virion/chemistry , Artificial Intelligence
11.
J Virol ; 98(2): e0189923, 2024 Feb 20.
Article En | MEDLINE | ID: mdl-38294245

After Epstein-Barr virus (EBV) genome replication and encapsidation in the nucleus, nucleocapsids are translocated into the cytoplasm for subsequent tegumentation and maturation. The EBV BGLF4 kinase, which induces partial disassembly of the nuclear lamina, and the nuclear egress complex BFRF1/BFLF2 coordinately facilitate the nuclear egress of nucleocapsids. Here, we demonstrate that within EBV reactivated epithelial cells, viral capsids, tegument proteins, and glycoproteins are clustered in the juxtanuclear concave region, accompanied by redistributed cytoplasmic organelles and the cytoskeleton regulator IQ-domain GTPase-activation protein 1 (IQGAP1), close to the microtubule-organizing center (MTOC). The assembly compartment (AC) structure was diminished in BGLF4-knockdown TW01-EBV cells and BGLF4-knockout bacmid-carrying TW01 cells, suggesting that the formation of AC structure is BGLF4-dependent. Notably, glycoprotein gp350/220 was observed by confocal imaging to be distributed in the perinuclear concave region and surrounded by the endoplasmic reticulum (ER) membrane marker calnexin, indicating that the AC may be located within a globular structure derived from ER membranes, adjacent to the outer nuclear membrane. Moreover, the viral capsid protein BcLF1 and tegument protein BBLF1 were co-localized with IQGAP1 near the cytoplasmic membrane in the late stage of replication. Knockdown of IQGAP1 did not affect the AC formation but decreased virion release from both TW01-EBV and Akata+ cells, suggesting IQGAP1-mediated trafficking regulates EBV virion release. The data presented here show that BGLF4 is required for cytoskeletal rearrangement, coordination with the redistribution of cytoplasmic organelles and IQGAP1 for virus maturation, and subsequent IQGAP1-dependent virion release.IMPORTANCEEBV genome is replicated and encapsidated in the nucleus, and the resultant nucleocapsids are translocated to the cytoplasm for subsequent virion maturation. We show that a cytoplasmic AC, containing viral proteins, markers of the endoplasmic reticulum, Golgi, and endosomes, is formed in the juxtanuclear region of epithelial and B cells during EBV reactivation. The viral BGLF4 kinase contributes to the formation of the AC. The cellular protein IQGAP1 is also recruited to the AC and partially co-localizes with the virus capsid protein BcLF1 and tegument protein BBLF1 in EBV-reactivated cells, dependent on the BGLF4-induced cytoskeletal rearrangement. In addition, virion release was attenuated in IQGAP1-knockdown epithelial and B cells after reactivation, suggesting that IQGAP1-mediated trafficking may regulate the efficiency of virus maturation and release.


Cytoplasm , Herpesvirus 4, Human , Protein Serine-Threonine Kinases , Viral Proteins , Virion , Virus Assembly , Virus Release , ras GTPase-Activating Proteins , Humans , Capsid Proteins/metabolism , Cytoplasm/metabolism , Cytoplasm/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , ras GTPase-Activating Proteins/metabolism , Viral Proteins/metabolism , Virion/chemistry , Virion/growth & development , Virion/metabolism , Virus Assembly/physiology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism
12.
Nucleic Acids Res ; 52(D1): D817-D821, 2024 Jan 05.
Article En | MEDLINE | ID: mdl-37897348

ViralZone (http://viralzone.expasy.org) is a knowledge repository for viruses that links biological knowledge and databases. It contains data on virion structure, genome, proteome, replication cycle and host-virus interactions. The new update provides better access to the data through contextual popups and higher resolution images in Scalable Vector Graphics (SVG) format. These images are designed to be dynamic and interactive with human viruses to give users better access to the data. In addition, a new coronavirus-specific resource provides regularly updated data on variants and molecular biology of SARS-CoV-2. Other virus-specific resources have been added to the database, particularly for HIV, herpesviruses and poxviruses.


Knowledge Bases , Viruses , Humans , Virion/chemistry , Virion/genetics , Virion/growth & development , Viruses/chemistry , Viruses/genetics , Viruses/growth & development
13.
Nature ; 623(7989): 1026-1033, 2023 Nov.
Article En | MEDLINE | ID: mdl-37993716

Human immunodeficiency virus 1 (HIV-1) infection is initiated by binding of the viral envelope glycoprotein (Env) to the cell-surface receptor CD41-4. Although high-resolution structures of Env in a complex with the soluble domains of CD4 have been determined, the binding process is less understood in native membranes5-13. Here we used cryo-electron tomography to monitor Env-CD4 interactions at the membrane-membrane interfaces formed between HIV-1 and CD4-presenting virus-like particles. Env-CD4 complexes organized into clusters and rings, bringing the opposing membranes closer together. Env-CD4 clustering was dependent on capsid maturation. Subtomogram averaging and classification revealed that Env bound to one, two and finally three CD4 molecules, after which Env adopted an open state. Our data indicate that asymmetric HIV-1 Env trimers bound to one and two CD4 molecules are detectable intermediates during virus binding to host cell membranes, which probably has consequences for antibody-mediated immune responses and vaccine immunogen design.


CD4 Antigens , Cell Membrane , HIV Envelope Protein gp120 , HIV-1 , Protein Multimerization , Humans , AIDS Vaccines/chemistry , AIDS Vaccines/immunology , Capsid/chemistry , Capsid/metabolism , Capsid/ultrastructure , CD4 Antigens/chemistry , CD4 Antigens/metabolism , CD4 Antigens/ultrastructure , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cryoelectron Microscopy , Electron Microscope Tomography , HIV Antibodies/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp120/ultrastructure , HIV Infections/virology , HIV-1/chemistry , HIV-1/ultrastructure , Virion/chemistry , Virion/metabolism , Virion/ultrastructure
14.
Soft Matter ; 19(44): 8649-8658, 2023 Nov 15.
Article En | MEDLINE | ID: mdl-37921635

Understanding the principles governing protein arrangement in viral capsids and structurally similar protein shells can enable the development of new antiviral strategies and the design of artificial protein cages for various applications. We study these principles within the context of the close packing problem, by analyzing dozens of small spherical shells assembled from a single type of protein. First, we use icosahedral spherical close packings containing 60T identical disks, where T ≤ 4, to rationalize the protein arrangement in twenty real icosahedral shells both satisfying and violating the paradigmatic Caspar-Klug model. We uncover a striking correspondence between the protein mass centers in the considered shells and the centers of disks in the close packings. To generalize the packing model, we consider proteins with a weak shape anisotropy and propose an interaction energy, minimization of which allows us to obtain spherical dense packings of slightly anisotropic structural units. In the case of strong anisotropy, we model the proteins as sequences of overlapping discs of different sizes, with minimum energy configuration not only resulting in packings, accurately reproducing locations and orientations of individual proteins, but also revealing that icosahedral packings that display the handedness of real capsids are energetically more favorable. Finally, by introducing effective disc charges, we rationalize the formation of inter-protein bonds in protein shells.


Capsid , Proteins , Capsid/chemistry , Virion/chemistry
15.
J Chem Theory Comput ; 19(21): 7924-7933, 2023 Nov 14.
Article En | MEDLINE | ID: mdl-37856311

For the first time, a complete all-atom molecular dynamics (MD) model of a virus, bacteriophage MS2, in its entirety, including a protein outer shell, native genomic RNA with necessary divalent ions, and surrounding explicit aqueous solution with ions at physiological concentration, was built. The model is based on an experimentally measured cryo-EM structure, which was substantially augmented by reconstructing missing or low-resolution parts of the measured density (where the atomistic structure cannot be fit unambiguously). The model was tested by a quarter of a microsecond MD run, and various biophysical characteristics are obtained and analyzed. The developed methodology of building the model can be used for reconstructing other large biomolecular structures when experimental data are fragmented and/or of varying resolution, while the model itself can be used for studying the biology of MS2, including the dynamics of its interaction with the host bacteria.


Levivirus , Molecular Dynamics Simulation , Levivirus/chemistry , Levivirus/genetics , Virion/chemistry , Ions , Cryoelectron Microscopy/methods
16.
J Virol ; 97(10): e0093823, 2023 10 31.
Article En | MEDLINE | ID: mdl-37792003

IMPORTANCE: Human norovirus (HuNoV) is highly infectious and can result in severe illnesses in the elderly and children. So far, there is no effective antiviral drug to treat HuNoV infection, and thus, the development of HuNoV vaccines is urgent. However, NoV evolves rapidly, and currently, at least 10 genogroups with numerous genotypes have been found. The genetic diversity of NoV and the lack of cross-protection between different genotypes pose challenges to the development of broadly protective vaccines. In this study, guided by structural alignment between GI.1 and GII.4 HuNoV VP1 proteins, several chimeric-type virus-like particles (VLPs) were designed through surface-exposed loop grafting. Mouse immunization studies show that two of the designed chimeric VLPs induced cross-immunity against both GI.1 and GII.4 HuNoVs. To our knowledge, this is the first designed chimeric VLPs that can induce cross-immune activities across different genogroups of HuNoV, which provides valuable strategies for the development of cross-reactive HuNoV vaccines.


Caliciviridae Infections , Epitopes , Genotype , Norovirus , Viral Vaccines , Virion , Animals , Humans , Mice , Caliciviridae Infections/immunology , Caliciviridae Infections/prevention & control , Caliciviridae Infections/virology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Immunization , Norovirus/chemistry , Norovirus/classification , Norovirus/genetics , Norovirus/immunology , Viral Vaccines/chemistry , Viral Vaccines/genetics , Viral Vaccines/immunology , Chimera/genetics , Chimera/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Virion/chemistry , Virion/genetics , Virion/immunology
17.
J Virol ; 97(11): e0081723, 2023 Nov 30.
Article En | MEDLINE | ID: mdl-37877717

IMPORTANCE: Understanding how bracoviruses (BVs) function in wasps is of broad interest in the study of virus evolution. This study characterizes most of the Microplitis demolitor bracovirus (MdBV) genes whose products are nucleocapsid components. Results indicate several genes unknown outside of nudiviruses and BVs are essential for normal capsid assembly. Results also indicate most MdBV tyrosine recombinase family members and the DNA binding protein p6.9-1 are required for DNA processing and packaging into nucleocapsids.


Capsid Proteins , Polydnaviridae , Virion , Animals , Capsid/chemistry , Capsid/metabolism , Polydnaviridae/genetics , Polydnaviridae/metabolism , Virion/chemistry , Virion/genetics , Virion/metabolism , Wasps/virology , Capsid Proteins/genetics , DNA-Binding Proteins/metabolism , Viral Genome Packaging , DNA, Viral/metabolism , Recombinases/metabolism
18.
J Virol ; 97(9): e0071023, 2023 09 28.
Article En | MEDLINE | ID: mdl-37681958

The envelope (Env) glycoproteins on HIV-1 virions are the sole target of broadly neutralizing antibodies (bNAbs) and the focus of vaccines. However, many cross-reactive conserved epitopes are often occluded on virus particles, contributing to the evasion of humoral immunity. This study aimed to identify the Env epitopes that are exposed/occluded on HIV-1 particles and to investigate the mechanisms contributing to their masking. Using a flow cytometry-based assay, three HIV-1 isolates, and a panel of antibodies, we show that only select epitopes, including V2i, the gp120-g41 interface, and gp41-MPER, are accessible on HIV-1 particles, while V3, V2q, and select CD4bs epitopes are masked. These epitopes become accessible after allosteric conformational changes are induced by the pre-binding of select Abs, prompting us to test if similar conformational changes are required for these Abs to exhibit their neutralization capability. We tested HIV-1 neutralization where the virus-mAb mix was pre-incubated/not pre-incubated for 1 hour prior to adding the target cells. Similar levels of neutralization were observed under both assay conditions, suggesting that the interaction between virus and target cells sensitizes the virions for neutralization via bNAbs. We further show that lectin-glycan interactions can also expose these epitopes. However, this effect is dependent on the lectin specificity. Given that, bNAbs are ideal for providing sterilizing immunity and are the goal of current HIV-1 vaccine efforts, these data offer insight on how HIV-1 may occlude these vulnerable epitopes from the host immune response. In addition, the findings can guide the formulation of effective antibody combinations for therapeutic use. IMPORTANCE The human immunodeficiency virus (HIV-1) envelope (Env) glycoprotein mediates viral entry and is the sole target of neutralizing antibodies. Our data suggest that antibody epitopes including V2q (e.g., PG9, PGT145), CD4bs (e.g., VRC01, 3BNC117), and V3 (2219, 2557) are masked on HIV-1 particles. The PG9 and 2219 epitopes became accessible for binding after conformational unmasking was induced by the pre-binding of select mAbs. Attempts to understand the masking mechanism led to the revelation that interaction between virus and host cells is needed to sensitize the virions for neutralization by broadly neutralizing antibodies (bNAbs). These data provide insight on how bNAbs may gain access to these occluded epitopes to exert their neutralization effects and block HIV-1 infection. These findings have important implications for the way we evaluate the neutralizing efficacy of antibodies and can potentially guide vaccine design.


Broadly Neutralizing Antibodies , Epitopes, B-Lymphocyte , HIV Antibodies , HIV Infections , HIV-1 , Host Microbial Interactions , Humans , Antibodies, Monoclonal/immunology , Broadly Neutralizing Antibodies/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/chemistry , HIV-1/immunology , HIV-1/metabolism , Lectins/metabolism , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , AIDS Vaccines/chemistry , AIDS Vaccines/immunology , Virion/chemistry , Virion/immunology , Virion/metabolism , Polysaccharides/metabolism
19.
ACS Sens ; 8(9): 3338-3348, 2023 09 22.
Article En | MEDLINE | ID: mdl-37610841

Our growing ability to tailor healthcare to the needs of individuals has the potential to transform clinical treatment. However, the measurement of multiple biomarkers to inform clinical decisions requires rapid, effective, and affordable diagnostics. Chronic diseases and rapidly evolving pathogens in a larger population have also escalated the need for improved diagnostic capabilities. Current chemical diagnostics are often performed in centralized facilities and are still dependent on multiple steps, molecular labeling, and detailed analysis, causing the result turnaround time to be over hours and days. Rapid diagnostic kits based on lateral flow devices can return results quickly but are only capable of detecting a handful of pathogens or markers. Herein, we present the use of disposable plasmonics with chiroptical nanostructures as a platform for low-cost, label-free optical biosensing with multiplexing and without the need for flow systems often required in current optical biosensors. We showcase the detection of SARS-CoV-2 in complex media as well as an assay for the Norovirus and Zika virus as an early developmental milestone toward high-throughput, single-step diagnostic kits for differential diagnosis of multiple respiratory viruses and any other emerging diagnostic needs. Diagnostics based on this platform, which we term "disposable plasmonics assays," would be suitable for low-cost screening of multiple pathogens or biomarkers in a near-point-of-care setting.


Biosensing Techniques , COVID-19 , Zika Virus Infection , Zika Virus , Humans , SARS-CoV-2 , COVID-19/diagnosis , Biosensing Techniques/methods , Virion/chemistry , Biomarkers/analysis
20.
Eur J Med Chem ; 259: 115578, 2023 Nov 05.
Article En | MEDLINE | ID: mdl-37467617

A synthetic multivalent hemagglutinin and neuraminidase inhibitor was developed by the conjugation of a septa-valent triazolylsialoside to bovine serum albumin using di-(N-succinimidyl) adipate. Matrixassisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) confirmed the attachment of five septa-valent sialyl lactosides to the protein backbone, resulting in a pentatrideca-valent sialyl conjugate. This pseudo-glycoprotein demonstrated a high affinity for hemagglutinin/neuraminidase as well as for the drug-resistant NA mutation on the influenza virus surface due to the cluster effect. The conjugate also exhibited potent antiviral activity against a wide range of virus strains without cytotoxicity at high concentrations. Mechanistic studies revealed that the pentatrideca-valent sialyl conjugate bound strongly to the influenza virion particles through interactions with HA/NA on the virion surfaces. The KD of the interaction was approximately 1 µM, as determined by isothermal calorimetric titration, allowing the capture and trapping of the influenza virions and preventing their further infection of host cells. These findings provide insight into the development of new antiviral agents using multivalent sialic acid clusters.


Influenza A virus , Influenza, Human , Humans , Hemagglutinins/analysis , Hemagglutinins/metabolism , Neuraminidase , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/genetics , Viral Proteins/metabolism , Virion/chemistry , Virion/genetics , Virion/metabolism
...