19,469 results on '"capsid proteins"'
Search Results
2. Characterization of Genetic Diversity in the Capsid Protein Gene of Grapevine Fleck Virus and Development of a New Real-Time RT-PCR Assay.
- Author
-
de Souza, Juliana, Klaassen, Vicki, Stevens, Kristian, Erickson, Teresa, Heinitz, Claire, and Al Rwahnih, Maher
- Subjects
RT-qPCR assay design ,capsid protein phylogeny ,grapevine ,grapevine fleck virus ,high throughput sequencing ,Capsid Proteins ,Genetic Variation ,Vitis ,Plant Diseases ,Phylogeny ,Real-Time Polymerase Chain Reaction ,Reverse Transcriptase Polymerase Chain Reaction ,Sequence Analysis ,DNA ,Flexiviridae - Abstract
The grapevine fleck virus (GFkV) is a ubiquitous grapevine-infecting virus found worldwide, is associated with the grapevine fleck complex, and is often found in mixed infections with viruses of the grapevine leafroll complex and/or vitiviruses. Although GFkV has been studied for a long time, limited sequence information is available in the public databases. In this study, the GFkV sequence data available in GenBank and data generated at the Foundation Plant Services, University of California, Davis, were used to perform nucleotide sequence comparisons, construct a phylogenetic tree, and develop a new RT-qPCR assay. Sequence comparisons showed high genetic diversity among the GFkV isolates, and the phylogenetic analyses revealed a new group comprised of GFkV isolates identified in the present study. A new assay, referred to as GFkV-CP, was designed and validated using an existing GFkV positive control together with 11 samples known to be infected with combinations of different marafiviruses and maculaviruses but not GFkV. In addition, the newly designed assay was used in a field survey to screen grapevines from diverse geographical locations that are maintained at the United States Department of Agriculture (USDA) National Clonal Germplasm Repository (NCGR) in Winters, CA.
- Published
- 2024
3. Systematic multi-trait AAV capsid engineering for efficient gene delivery.
- Author
-
Eid, Fatma-Elzahraa, Chen, Albert, Chan, Ken, Huang, Qin, Zheng, Qingxia, Tobey, Isabelle, Pacouret, Simon, Brauer, Pamela, Keyes, Casey, Powell, Megan, Johnston, Jencilin, Zhao, Binhui, Lage, Kasper, Tarantal, Alice, Chan, Yujia, and Deverman, Benjamin
- Subjects
Dependovirus ,Animals ,Humans ,Mice ,Genetic Vectors ,Capsid ,Capsid Proteins ,Liver ,Transduction ,Genetic ,Gene Transfer Techniques ,Machine Learning ,Genetic Therapy ,Macaca ,Hepatocytes ,HEK293 Cells ,Genetic Engineering - Abstract
Broadening gene therapy applications requires manufacturable vectors that efficiently transduce target cells in humans and preclinical models. Conventional selections of adeno-associated virus (AAV) capsid libraries are inefficient at searching the vast sequence space for the small fraction of vectors possessing multiple traits essential for clinical translation. Here, we present Fit4Function, a generalizable machine learning (ML) approach for systematically engineering multi-trait AAV capsids. By leveraging a capsid library that uniformly samples the manufacturable sequence space, reproducible screening data are generated to train accurate sequence-to-function models. Combining six models, we designed a multi-trait (liver-targeted, manufacturable) capsid library and validated 88% of library variants on all six predetermined criteria. Furthermore, the models, trained only on mouse in vivo and human in vitro Fit4Function data, accurately predicted AAV capsid variant biodistribution in macaque. Top candidates exhibited production yields comparable to AAV9, efficient murine liver transduction, up to 1000-fold greater human hepatocyte transduction, and increased enrichment relative to AAV9 in a screen for liver transduction in macaques. The Fit4Function strategy ultimately makes it possible to predict cross-species traits of peptide-modified AAV capsids and is a critical step toward assembling an ML atlas that predicts AAV capsid performance across dozens of traits.
- Published
- 2024
4. Mouse and human immune responses share neutralization epitopes of HAstV-VA1
- Author
-
Ramírez-Bello, Inci, López, Tomás, Espinosa, Rafaela, Ghosh, Anisa, Green, Kassidy, Riaño-Umbarila, Lidia, Gaspar-Castillo, Carlos, Aguilera-Flores, Catalina, Alpuche-Aranda, Celia M, López, Susana, DuBois, Rebecca M, and Arias, Carlos F
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Immunization ,Vaccine Related ,Biotechnology ,Infectious Diseases ,2.1 Biological and endogenous factors ,5.2 Cellular and gene therapies ,Infection ,Humans ,Animals ,Antibodies ,Neutralizing ,Mice ,Epitopes ,Antibodies ,Viral ,Antibodies ,Monoclonal ,Capsid Proteins ,Mamastrovirus ,Mutation ,Astroviridae Infections ,Neutralization Tests ,VA1 astrovirus ,neutralizing monoclonal antibody ,epitope mapping ,immunodeficient patient ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
Astroviruses are highly divergent and infect a wide variety of animal hosts. In 2009, a genetically divergent human astrovirus (HAstV) strain VA1 was first identified in an outbreak of acute gastroenteritis. This strain has also been associated with fatal central nervous system disease. In this work, we report the isolation of three high-affinity neutralizing monoclonal antibodies (Nt-MAbs) targeting the capsid spike domain of HAstV-VA1. These antibodies (7C8, 2A2, 3D8) were used to select individual HAstV-VA1 mutants resistant to their neutralizing activity and a HAstV-VA1 triple mutant that escapes neutralization from all three Nt-MAbs. Sequencing of the virus genome capsid region revealed escape mutations that map to the surface of the capsid spike domain, define three potentially independent neutralization epitopes, and help delineate four antigenic sites in human astroviruses. Notably, two of the escape mutations were found to be present in the spike sequence of the HAstV-VA1-PS strain isolated from an immunodeficient patient with encephalitis, suggesting that those mutations arose as a result of the immune pressure generated by the patient's immunotherapy. In agreement with this observation, human serum samples exhibiting strong neutralization activity against wild-type HAstV-VA1 had a 2.6-fold reduction in neutralization titer when evaluated against the triple-escape HAstV-VA1 mutant, suggesting that both mouse and human antibody responses target shared neutralization epitopes. The isolated Nt-MAbs reported in this work will help to characterize the functional domains of the virus during cell entry and have the potential for developing a specific antibody therapy for the neurological disease associated with HAstV-VA1.ImportanceHuman astroviruses (HAstVs) have been historically associated with acute gastroenteritis. However, the genetically divergent HAstV-VA1 strain has been associated with central nervous system disease. In this work high-affinity neutralizing monoclonal antibodies directed to HAstV-VA1 were isolated and characterized. The proposed binding sites for these antibodies and for neutralizing antibodies against classical HAstVs suggest that there are at least four neutralization sites on the capsid spike of astroviruses. Our data show that natural infection with human astrovirus VA1 elicits a robust humoral immune response that targets the same antigenic sites recognized by the mouse monoclonal antibodies and strongly suggests the emergence of a variant HAstV-VA1 virus in an immunodeficient patient with prolonged astrovirus infection. The isolated Nt-MAb reported in this work will help to define the functional sites of the virus involved in cell entry and hold promise for developing a specific antibody therapy for the neurological disease associated with HAstV-VA1.
- Published
- 2024
5. Capsid-dependent lentiviral restrictions.
- Author
-
Twentyman, Joy, Emerman, Michael, and Ohainle, Molly
- Subjects
capsid ,human immunodeficiency virus ,lentiviruses ,restriction factor ,Animals ,Capsid ,Capsid Proteins ,Lentivirus ,Host-Pathogen Interactions ,Lentivirus Infections - Abstract
Host antiviral proteins inhibit primate lentiviruses and other retroviruses by targeting many features of the viral life cycle. The lentiviral capsid protein and the assembled viral core are known to be inhibited through multiple, directly acting antiviral proteins. Several phenotypes, including those known as Lv1 through Lv5, have been described as cell type-specific blocks to infection against some but not all primate lentiviruses. Here we review important features of known capsid-targeting blocks to infection together with several blocks to infection for which the genes responsible for the inhibition still remain to be identified. We outline the features of these blocks as well as how current methodologies are now well suited to find these antiviral genes and solve these long-standing mysteries in the HIV and retrovirology fields.
- Published
- 2024
6. Divergent Effects of Circoviridae Capsid Proteins on Type I Interferon Signaling.
- Author
-
Kosaka, Anon H., Huang, Chen-Yu, Lu, Zih-Ying, Hsing, Hua-Zhen, Choonnasard, Amonrat, Ringo, Rissar Siringo, Chuang, Kuo Pin, and Saito, Akatsuki
- Subjects
PSITTACINE beak & feather disease ,MOLECULAR mechanisms of immunosuppression ,WASTING syndrome ,VIRUS diseases ,RESPIRATORY diseases - Abstract
Viruses in the Circoviridae family can infect mammals and birds. Porcine circovirus type 2 (PCV2) significantly affects the livestock industry by causing porcine circovirus-associated diseases, such as postweaning multisystem wasting syndrome, respiratory disease complex, and dermatitis nephropathy syndrome. Additionally, beak and feather disease virus in parrots, canine circovirus in dogs, and columbid circovirus (pigeon circovirus) in racing pigeons induce immunosuppression, followed by secondary infections in these hosts. Although the PCV2 capsid protein has been demonstrated to inhibit type I interferon (IFN) signaling, the molecular mechanisms of Circoviridae-induced immunosuppression are largely unknown. In this study, we examined whether these functions are conserved across Circoviridae capsid proteins. Our results illustrated that although the nuclear localization of capsid proteins is conserved, their effects on IFN-β signaling vary by species, revealing the diverse roles of Circoviridae capsid proteins in modulating immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
7. A new scheme for boosting in situ Fenton-like reaction in plant pathogenic tissues for selective structural degradation of capsid proteins
- Author
-
Jun Ma, Ming Chen, Yujie Wang, Jiaying Li, Jian Tang, Shengjun Wu, Chi He, and J. Paul Chen
- Subjects
Internal doping ,Fenton-like reactions ,Plant viruses ,Capsid proteins ,Biotechnology ,TP248.13-248.65 ,Medical technology ,R855-855.5 - Abstract
Abstract Safe prevention and control of plant viruses is a global challenge. Inducing viral capsid protein (CP) degradation via hydroxyl radicals (∙OH) generated by an in situ Fenton-like reaction within plant pathogenic tissues is proposed for combating plant viruses in this study. We designed a new Fenton-like reaction inducer, tCuinter-bCDs, which utilizes an internal doping strategy that reduces copper content by 89.89% compared to conventional doping methods, while still achieving a high coexistence of multivalent copper ions. Our research demonstrated that tCuinter-bCDs possessed functional activity to specifically recognize and proximally degrade CP. tCuinter-bCDs form complexes with CP monomers through supramolecular bonds characterized by significant electrostatic components. Within 10 min, the complex induced complete degradation of tertiary structure pockets composed of α-helices and β-sheets located at residues MET1-GLU23, TYR73-ARG93, and SER147-PRO157. Based on a high-resolution 2.91 Å CP model that was constructed for the first time, this degradation process is likely driven by hydrophobic interactions between tCuinter-bCDs and CP residues MET1, VAL5, THR55, and THR58, along with hydrogen bonds formed with THR4, VAL5, GLY15, PRO57, and ALA59, thereby promoting degradation of adjacent peptide segments. This represents the first study demonstrating in situ Fenton-like reactions to combat pathogens in plant systems. Our findings provide a new, efficient, and environmentally friendly approach for plant virus control. Graphical Abstract
- Published
- 2025
- Full Text
- View/download PDF
8. The incredible bulk: Human cytomegalovirus tegument architectures uncovered by AI-empowered cryo-EM
- Author
-
Jih, Jonathan, Liu, Yun-Tao, Liu, Wei, and Zhou, Z Hong
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,2.2 Factors relating to the physical environment ,Generic health relevance ,Humans ,Cytomegalovirus ,Cryoelectron Microscopy ,Capsid Proteins ,Capsid ,Virion ,Artificial Intelligence - Abstract
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.
- Published
- 2024
9. Structure and Antigenicity of the Porcine Astrovirus 4 Capsid Spike
- Author
-
Haley, Danielle J, Lanning, Sarah, Henricson, Kyle E, Mardirossian, Andre A, Cirillo, Iyan, Rahe, Michael C, and DuBois, Rebecca M
- Subjects
Microbiology ,Biological Sciences ,Infectious Diseases ,Biotechnology ,Immunization ,Vaccine Related ,Infection ,Good Health and Well Being ,Animals ,Swine ,Capsid Proteins ,Swine Diseases ,Antibodies ,Viral ,Astroviridae Infections ,Antigens ,Viral ,Capsid ,Recombinant Proteins ,Epitopes ,Models ,Molecular ,Viral Vaccines ,Protein Conformation ,Mamastrovirus ,astroviruses ,virus structure ,spike ,porcine astrovirus ,capsid - Abstract
Porcine astrovirus 4 (PoAstV4) has been recently associated with respiratory disease in pigs. In order to understand the scope of PoAstV4 infections and to support the development of a vaccine to combat PoAstV4 disease in pigs, we designed and produced a recombinant PoAstV4 capsid spike protein for use as an antigen in serological assays and for potential future use as a vaccine antigen. Structural prediction of the full-length PoAstV4 capsid protein guided the design of the recombinant PoAstV4 capsid spike domain expression plasmid. The recombinant PoAstV4 capsid spike was expressed in Escherichia coli, purified by affinity and size-exclusion chromatography, and its crystal structure was determined at 1.85 Å resolution, enabling structural comparisons to other animal and human astrovirus capsid spike structures. The recombinant PoAstV4 capsid spike protein was also used as an antigen for the successful development of a serological assay to detect PoAstV4 antibodies, demonstrating that the recombinant PoAstV4 capsid spike retains antigenic epitopes found on the native PoAstV4 capsid. These studies lay a foundation for seroprevalence studies and the development of a PoAstV4 vaccine for swine.
- Published
- 2024
10. Structure and antigenicity of the divergent human astrovirus VA1 capsid spike
- Author
-
Ghosh, Anisa, Delgado-Cunningham, Kevin, López, Tomás, Green, Kassidy, Arias, Carlos F, and DuBois, Rebecca M
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Infectious Diseases ,Digestive Diseases ,Foodborne Illness ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Infection ,Child ,Humans ,Capsid ,Mamastrovirus ,Capsid Proteins ,Mutation ,Astroviridae Infections ,Phylogeny ,Feces ,Microbiology ,Immunology ,Virology ,Medical microbiology - Abstract
Human astrovirus (HAstV) is a known cause of viral gastroenteritis in children worldwide, but HAstV can cause also severe and systemic infections in immunocompromised patients. There are three clades of HAstV: classical, MLB, and VA/HMO. While all three clades are found in gastrointestinal samples, HAstV-VA/HMO is the main clade associated with meningitis and encephalitis in immunocompromised patients. To understand how the HAstV-VA/HMO can infect the central nervous system, we investigated its sequence-divergent capsid spike, which functions in cell attachment and may influence viral tropism. Here we report the high-resolution crystal structures of the HAstV-VA1 capsid spike from strains isolated from patients with gastrointestinal and neuronal disease. The HAstV-VA1 spike forms a dimer and shares a core beta-barrel structure with other astrovirus capsid spikes but is otherwise strikingly different, suggesting that HAstV-VA1 may utilize a different cell receptor, and an infection competition assay supports this hypothesis. Furthermore, by mapping the capsid protease cleavage site onto the structure, the maturation and assembly of the HAstV-VA1 capsid is revealed. Finally, comparison of gastrointestinal and neuronal HAstV-VA1 sequences, structures, and antigenicity suggests that neuronal HAstV-VA1 strains may have acquired immune escape mutations. Overall, our studies on the HAstV-VA1 capsid spike lay a foundation to further investigate the biology of HAstV-VA/HMO and to develop vaccines and therapeutics targeting it.
- Published
- 2024
11. Human papillomavirus vaccines: organisation and experience of preclinical studies
- Author
-
A. S. Korovkin, T. N. Nikitina, T. Yu. Kozlova, D. V. Gorenkov, and A. R. Volgin
- Subjects
papillomavirus vaccines ,papillomavirus infections ,hpv vaccines ,vaccination ,capsid proteins ,l1 protein ,adjuvants ,hpv serotypes ,animals ,preclinical studies ,Biotechnology ,TP248.13-248.65 ,Medicine - Abstract
INTRODUCTION. Vaccination is the main measure for the primary prevention of human papillomavirus (HPV)-related diseases. The development of novel vaccine candidates is underway worldwide, including in the Russian Federation. At the same time, the clinical introduction of new HPV vaccines is seriously hampered by the lack of clear and unambiguous recommendations for conducting preclinical studies of these vaccines.AIM. This study aimed to analyse regulatory documents on HPV vaccines, to study the experience of conducting preclinical studies, and to summarise the preclinical approaches that could be recommended for developers and applicants seeking approval for new preventive HPV vaccines, including the vaccines being developed in the Russian Federation.DISCUSSION. The authors have analysed regulatory documents issued by the World Health Organisation (WHO), the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), and the Council of the Eurasian Economic Commission (EEC). Additionally, the authors have reviewed the experimental results of preclinical studies of HPV vaccines. The known licensed and pipeline HPV vaccines are similar in terms of their characteristics and constructive features. However, there may be some differences in the HPV serotype coverage and the methods used to produce the HPV L1 capsid protein. To date, studies have confirmed the role of the HPV L1 capsid protein in the development of specific immunity, rendering challenge tests in animal models unnecessary. Papillomatosis modelling may be required for choosing an alternative immunological target or for studying an alternative (non-parenteral) route for vaccine administration. Preclinical study programmes may be supplemented with individual stages of comprehensive assessment of adjuvants and other additives included in novel HPV vaccine compositions.CONCLUSIONS. The authors have studied the international experience and presented a systemic overview of the methods and approaches used in preclinical studies of HPV vaccines. The authors have formulated recommendations for developers for the planning and organisation of preclinical studies of HPV vaccines (including immunogenicity, toxicity, and local tolerance assessments required for licensing new vaccines).
- Published
- 2024
- Full Text
- View/download PDF
12. Structure and immunogenicity of the murine astrovirus capsid spike.
- Author
-
Lanning, Sarah, Pedicino, Natalie, Haley, Danielle J, Hernandez, Samuel, Cortez, Valerie, and DuBois, Rebecca M
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biotechnology ,Infectious Diseases ,Immunization ,Prevention ,Vaccine Related ,3.4 Vaccines ,Infection ,Good Health and Well Being ,Child ,Humans ,Animals ,Mice ,Child ,Preschool ,Capsid ,Capsid Proteins ,Astroviridae ,Astroviridae Infections ,Vaccines ,astroviruses ,virus structure ,spike protein ,murine astrovirus ,viral capsid. ,viral capsid ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
Human astroviruses (HAstVs) are small, non-enveloped icosahedral RNA viruses that are a significant cause of diarrhoea in young children. Despite their worldwide prevalence, HAstV pathogenesis studies and vaccine development remain challenging due to the lack of an animal model for HAstV infection. The recent development of a murine astrovirus (MuAstV) infection model in mice provides the opportunity to test proof-of-concept vaccines based on MuAstV antigens. To help establish a system in which an astrovirus capsid spike-based vaccine could be tested in vivo, we designed and produced a recombinant MuAstV capsid spike protein based on predicted secondary structure homology to HAstV spike proteins. The recombinant MuAstV spike can be expressed with high efficiency in Escherichia coli and retains antigenicity to polyclonal antibodies elicited by MuAstV infection. We determined the crystal structure of the MuAstV spike to 1.75 Å and assessed its structural conservation with HAstV capsid spike. Despite low sequence identity between the MuAstV and HAstV spikes and differences in their overall shapes, they share related structural folds. Additionally, we found that vaccination with MuAstV spike induced anti-MuAstV-spike antibodies, highlighting that the recombinant spike is immunogenic. These studies lay a foundation for future in vivo MuAstV challenge studies to test whether MuAstV spike can be the basis of an effective vaccine.
- Published
- 2023
13. Identification of the bacteriophage nucleus protein interaction network.
- Author
-
Deep, Amar, Gu, Yajie, Nguyen, Katrina, Chaikeeratisak, Vorrapon, Armbruster, Emily, Ghassemian, Majid, Corbett, Kevin, Pogliano, Joseph, Villa, Elizabeth, and Enustun, Eray
- Subjects
Bacteriophages ,Protein Interaction Maps ,Capsid ,Capsid Proteins ,RNA ,Messenger - Abstract
In the arms race between bacteria and bacteriophages (phages), some large-genome jumbo phages have evolved a protein shell that encloses their replicating genome to protect it against host immune factors. By segregating the genome from the host cytoplasm, however, the phage nucleus introduces the need to specifically translocate messenger RNA and proteins through the nuclear shell and to dock capsids on the shell for genome packaging. Here, we use proximity labeling and localization mapping to systematically identify proteins associated with the major nuclear shell protein chimallin (ChmA) and other distinctive structures assembled by these phages. We identify six uncharacterized nuclear-shell-associated proteins, one of which directly interacts with self-assembled ChmA. The structure and protein-protein interaction network of this protein, which we term ChmB, suggest that it forms pores in the ChmA lattice that serve as docking sites for capsid genome packaging and may also participate in messenger RNA and/or protein translocation.
- Published
- 2023
14. Antiviral Agents: Structural Basis of Action and Rational Design
- Author
-
Menéndez-Arias, Luis, Gago, Federico, Kundu, Tapas K., Series Editor, Harris, J. Robin, Advisory Editor, Holzenburg, Andreas, Advisory Editor, Korolchuk, Viktor, Advisory Editor, Bolanos-Garcia, Victor, Advisory Editor, Marles-Wright, Jon, Advisory Editor, and Mateu, Mauricio G., editor
- Published
- 2024
- Full Text
- View/download PDF
15. Characterizing Heterogeneous Mixtures of Assembled States of the Tobacco Mosaic Virus Using Charge Detection Mass Spectrometry
- Author
-
Bischoff, Amanda J, Harper, Conner C, Williams, Evan R, and Francis, Matthew B
- Subjects
Analytical Chemistry ,Chemical Sciences ,Physical Chemistry ,Biotechnology ,Tobacco Mosaic Virus ,Capsid Proteins ,Chemical Phenomena ,General Chemistry ,Chemical sciences ,Engineering - Abstract
The tobacco mosaic viral capsid protein (TMV) is a frequent target for derivatization for myriad applications, including drug delivery, biosensing, and light harvesting. However, solutions of the stacked disk assembly state of TMV are difficult to characterize quantitatively due to their large size and multiple assembled states. Charge detection mass spectrometry (CDMS) addresses the need to characterize heterogeneous populations of large protein complexes in solution quickly and accurately. Using CDMS, previously unobserved assembly states of TMV, including 16-monomer disks and odd-numbered disk stacks, have been characterized. We additionally employed a peptide-protein conjugation reaction in conjunction with CDMS to demonstrate that modified TMV proteins do not redistribute between disks. Finally, this technique was used to discriminate between protein complexes of near-identical mass but different configurations. We have gained a greater understanding of the behavior of TMV, a protein used across a broad variety of fields and applications, in the solution state.
- Published
- 2022
16. Fitness Landscape-Guided Engineering of Locally Supercharged Virus-like Particles with Enhanced Cell Uptake Properties
- Author
-
Pistono, Paige E, Huang, Paul, Brauer, Daniel D, and Francis, Matthew B
- Subjects
Biotechnology ,Bioengineering ,Nanotechnology ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Generic health relevance ,Animals ,Capsid ,Capsid Proteins ,Nanoparticles ,Drug Delivery Systems ,Mammals ,Chemical Sciences ,Biological Sciences ,Organic Chemistry - Abstract
Protein-based nanoparticles are useful models for the study of self-assembly and attractive candidates for drug delivery. Virus-like particles (VLPs) are especially promising platforms for expanding the repertoire of therapeutics that can be delivered effectively as they can deliver many copies of a molecule per particle for each delivery event. However, their use is often limited due to poor uptake of VLPs into mammalian cells. In this study, we use the fitness landscape of the bacteriophage MS2 VLP as a guide to engineer capsid variants with positively charged surface residues to enhance their uptake into mammalian cells. By combining mutations with positive fitness scores that were likely to produce assembled capsids, we identified two key double mutants with internalization efficiencies as much as 67-fold higher than that of wtMS2. Internalization of these variants with positively charged surface residues depends on interactions with cell surface sulfated proteoglycans, and yet, they are biophysically similar to wtMS2 with low cytotoxicity and an overall negative charge. Additionally, the best-performing engineered MS2 capsids can deliver a potent anticancer small-molecule therapeutic with efficacy levels similar to antibody-drug conjugates. Through this work, we were able to establish fitness landscape-based engineering as a successful method for designing VLPs with improved cell penetration. These findings suggest that VLPs with positive surface charge could be useful in improving the delivery of small-molecule- and nucleic acid-based therapeutics.
- Published
- 2022
17. Structure of the divergent human astrovirus MLB capsid spike
- Author
-
Delgado-Cunningham, Kevin, López, Tomás, Khatib, Firas, Arias, Carlos F, and DuBois, Rebecca M
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Machine Learning and Artificial Intelligence ,2.2 Factors relating to the physical environment ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Humans ,Capsid ,Mamastrovirus ,Capsid Proteins ,Binding Sites ,Machine Learning ,capsid ,crystal structure ,human astrovirus ,protein structure prediction ,Chemical Sciences ,Information and Computing Sciences ,Biophysics ,Biological sciences ,Chemical sciences - Abstract
Despite their worldwide prevalence and association with human disease, the molecular bases of human astrovirus (HAstV) infection and evolution remain poorly characterized. Here, we report the structure of the capsid protein spike of the divergent HAstV MLB clade (HAstV MLB). While the structure shares a similar folding topology with that of classical-clade HAstV spikes, it is otherwise strikingly different. We find no evidence of a conserved receptor-binding site between the MLB and classical HAstV spikes, suggesting that MLB and classical HAstVs utilize different receptors for host-cell attachment. We provide evidence for this hypothesis using a novel HAstV infection competition assay. Comparisons of the HAstV MLB spike structure with structures predicted from its sequence reveal poor matches, but template-based predictions were surprisingly accurate relative to machine-learning-based predictions. Our data provide a foundation for understanding the mechanisms of infection by diverse HAstVs and can support structure determination in similarly unstudied systems.
- Published
- 2022
18. Divergent Effects of Circoviridae Capsid Proteins on Type I Interferon Signaling
- Author
-
Anon H. Kosaka, Chen-Yu Huang, Zih-Ying Lu, Hua-Zhen Hsing, Amonrat Choonnasard, Rissar Siringo Ringo, Kuo Pin Chuang, and Akatsuki Saito
- Subjects
Circoviridae ,capsid proteins ,interferon β signaling ,Medicine - Abstract
Viruses in the Circoviridae family can infect mammals and birds. Porcine circovirus type 2 (PCV2) significantly affects the livestock industry by causing porcine circovirus-associated diseases, such as postweaning multisystem wasting syndrome, respiratory disease complex, and dermatitis nephropathy syndrome. Additionally, beak and feather disease virus in parrots, canine circovirus in dogs, and columbid circovirus (pigeon circovirus) in racing pigeons induce immunosuppression, followed by secondary infections in these hosts. Although the PCV2 capsid protein has been demonstrated to inhibit type I interferon (IFN) signaling, the molecular mechanisms of Circoviridae-induced immunosuppression are largely unknown. In this study, we examined whether these functions are conserved across Circoviridae capsid proteins. Our results illustrated that although the nuclear localization of capsid proteins is conserved, their effects on IFN-β signaling vary by species, revealing the diverse roles of Circoviridae capsid proteins in modulating immune responses.
- Published
- 2025
- Full Text
- View/download PDF
19. DNA Origami Disguises Herpes Simplex Virus 1 Particles and Controls Their Virulence.
- Author
-
Borum, Raina, Lin, Avery, Dong, Xiangyi, Kai, Mingxuan, and Chen, Yi
- Subjects
DNA origami ,folic acid ,herpes simplex virus ,targeted delivery ,Humans ,Herpesvirus 1 ,Human ,Virulence ,Capsid ,DNA ,Capsid Proteins ,Herpes Simplex - Abstract
DNA nanostructures are well-established vectors for packaging diversified payloads for targeted cellular delivery. Here, DNA origami rectangular sheets were combined with Herpes Simplex Virus 1 (HSV1) capsids to demonstrate surface coverage of the particle via electrostatic interactions. The optimized origami:HSV1 molar ratios led to characteristic packaging geometries ranging from dispersed HSV1 pockets to agglomerated HSV1 sleeves. Pockets were disguised from cells in HeLa and B16F10 cells and were 44.2% less infective than naked HSV1 particles. However, the pockets were 117% more infective than naked HSV1 particles when the origami sheets were coated with folic acid. We observed infectivity from naked origami, but they are 99.1% less infective with respect to HSV1 and 99.6% less infective with respect to the pocket complexes. This work suggests that DNA origami can selectively modulate virus infectivity.
- Published
- 2022
20. Characterizing Glycosylation of Adeno-Associated Virus Serotype 9 Capsid Proteins Generated from HEK293 Cells through Glycopeptide Mapping and Released Glycan Analysis.
- Author
-
Zhou, Yu, Priya, Sonal, and Ong, Joseph Y.
- Subjects
ADENO-associated virus ,LIQUID chromatography-mass spectrometry ,GLYCANS ,GLYCOSYLATION ,HYDROPHILIC interactions ,GENE therapy ,BOTULINUM A toxins - Abstract
Recombinant adeno-associated viral (AAV) vectors have emerged as prominent gene delivery vehicles for gene therapy. AAV capsid proteins determine tissue specificity and immunogenicity and play important roles in receptor binding, the escape of the virus from the endosome, and the transport of the viral DNA to the nuclei of target cells. Therefore, the comprehensive characterization of AAV capsid proteins is necessary for a better understanding of the vector assembly, stability, and transduction efficiency of AAV gene therapies. Glycosylation is one of the most common post-translational modifications (PTMs) and may affect the tissue tropism of AAV gene therapy. However, there are few studies on the characterization of the N- and O-glycosylation of AAV capsid proteins. In this study, we identified the N- and O-glycosylation sites and forms of AAV9 capsid proteins generated from HEK293 cells using liquid chromatography–tandem mass spectrometry (LC-MS)-based glycopeptide mapping and identified free N-glycans released from AAV9 capsid proteins by PNGase F using hydrophilic interaction (HILIC) LC-MS and HILIC LC-fluorescence detection (FLD) methods. This study demonstrates that AAV9 capsids are sprinkled with sugars, including N- and O-glycans, albeit at low levels. It may provide valuable information for a better understanding of AAV capsids in supporting AAV-based gene therapy development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Characterization of Adeno-Associated Virus Capsid Proteins by Microflow Liquid Chromatography Coupled with Mass Spectrometry.
- Author
-
Qin, Xi, Li, Xiang, Chen, Lingsheng, Gao, Tie, Luo, Ji, Guo, Lihai, Mollah, Sahana, Zhang, Zoe, Zhou, Yong, and Chen, Hong-Xu
- Abstract
Adeno-associated virus (AAV) has been widely used to treat various human diseases as an important delivery vector for gene therapy due to its low immunogenicity and safety. AAV capsids proteins are comprised of three capsid viral proteins (VP; VP1, VP2, VP3). The capsid proteins play a key role in viral vector infectivity and transduction efficiency. To ensure the safety and efficacy of AAV gene therapy products, the quality of AAV vector capsid proteins during development and production should be carefully monitored and controlled. Microflow liquid chromatography coupled with mass spectrometry provides superior sensitivity and fast analysis capability. It showed significant advantages in the analysis of low- concentration and large numbers of AAV samples. The intact mass of capsid protein can be accurately determined using high-resolution mass spectrometry (MS). And MS also provides highly confident confirmation of sequence coverage and post-translational modifications site identification and quantitation. In this study, we used microflow liquid chromatography-tandem mass spectrometry (LC–MS/MS) for the characterization of AAV2 capsid protein. we obtained nearly 100% sequence coverage of low-concentration AAV2 capsid protein (8 × 10
11 GC/mL). More than 30 post-translational modifications (PTMs) sites were identified, the PTMs types included deamidation, oxidation and acetylation. From this study, the proposed microflow LC–MS/MS method provides a sensitive and high throughput approach in the characterization of AAVs and other biological products with low abundance. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
22. The Capsid Precursor Protein of Astrovirus VA1 Is Proteolytically Processed Intracellularly
- Author
-
Aguilera-Flores, Catalina, López, Tomás, Zamudio, Fernando, Sandoval-Jaime, Carlos, Pérez, Edmundo I, López, Susana, DuBois, Rebecca, and Arias, Carlos F
- Subjects
Infectious Diseases ,2.2 Factors relating to the physical environment ,Aetiology ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,Astroviridae Infections ,Caco-2 Cells ,Capsid ,Capsid Proteins ,Humans ,Intracellular Space ,Mamastrovirus ,Protein Precursors ,Trypsin ,VA1 astrovirus ,proteolytical cleavage ,RNA virus ,capsid processing ,plus-strand RNA virus ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology - Abstract
Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients, and its recent propagation in cell culture has opened the possibility to study its biology. Unlike classical human astroviruses, VA1 growth was found to be independent of trypsin during virus replication in vitro. In this work, we show that despite its independence on trypsin activation for cell infection, the VA1 capsid precursor protein, of 86 kDa (VP86), is processed intracellularly, and this proteolytic processing is important for astrovirus VA1 infectivity. Antibodies raised against different regions of the capsid precursor showed that the polyprotein can be processed starting at either its amino- or carboxy-terminal end, and they allowed us to identify those proteins of about 33 (VP33) and 38 (VP38) kDa constitute the core and the spike proteins of the mature infectious virus particles, respectively. The amino-terminal end of the spike protein was found to be Thr-348. Whether the protease involved in intracellular cleavage of the capsid precursor is of viral or cellular origin remains to be determined, but the cleavage is independent of caspases. Also, trypsin is able to degrade the capsid precursor but has no effect on VP33 and VP38 proteins when assembled into virus particles. These studies provide the basis for advancement of the knowledge of astrovirus VA1 cell entry and replication. IMPORTANCE Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients. Its recent propagation in cell culture has facilitated the study of its biology. In this work, we show that despite the ability of this virus to grow in the absence of trypsin, a marked feature of human classical astroviruses, the capsid precursor protein of astrovirus VA1 is cleaved intracellularly to yield the mature infectious particles, formed by two polypeptides, VP33 that constitutes the core domain of the virus particle, and VP38 that forms the spike of the virus. These studies provide a platform to advance our knowledge on astrovirus VA1 cell entry and replication.
- Published
- 2022
23. Dissolving Microneedle Delivery of a Prophylactic HPV Vaccine
- Author
-
Ray, Sayoni, Wirth, David M, Ortega-Rivera, Oscar A, Steinmetz, Nicole F, and Pokorski, Jonathan K
- Subjects
Sexually Transmitted Infections ,Cancer ,HIV/AIDS ,Infectious Diseases ,Immunization ,Vaccine Related ,HPV and/or Cervical Cancer Vaccines ,Biotechnology ,Prevention ,Cervical Cancer ,Prevention of disease and conditions ,and promotion of well-being ,3.4 Vaccines ,Infection ,Good Health and Well Being ,Animals ,Antibodies ,Viral ,Capsid Proteins ,Epitopes ,Humans ,Mice ,Mice ,Inbred BALB C ,Papillomavirus Infections ,Papillomavirus Vaccines ,Vaccines ,Virus-Like Particle ,Chemical Sciences ,Biological Sciences ,Engineering ,Polymers - Abstract
Prophylactic vaccines capable of preventing human papillomavirus (HPV) infections are still inaccessible to a vast majority of the global population due to their high cost and challenges related to multiple administrations performed in a medical setting. In an effort to improve distribution and administration, we have developed dissolvable microneedles loaded with a thermally stable HPV vaccine candidate consisting of Qβ virus-like particles (VLPs) displaying a highly conserved epitope from the L2 protein of HPV (Qβ-HPV). Polymeric microneedle delivery of Qβ-HPV produces similar amounts of anti-HPV16 L2 IgG antibodies compared to traditional subcutaneous injection while delivering a much smaller amount of intradermal dose. However, a dose sparing effect was found. Furthermore, immunization yielded neutralizing antibody responses in a HPV pseudovirus assay. The vaccine candidate was confirmed to be stable at room temperature after storage for several months, potentially mitigating many of the challenges associated with cold-chain distribution. The ease of self-administration and minimal invasiveness of such microneedle patch vaccines may enable wide-scale distribution of the HPV vaccine and lead to higher patient compliance. The Qβ VLP and its delivery technology is a plug-and-play system that could serve as a universal platform with a broad range of applications. Qβ VLPs may be stockpiled for conjugation to a wide range of epitopes, which are then packaged and delivered directly to the patient via noninvasive microneedle patches. Such a system paves the way for rapid distribution and self-administration of vaccines.
- Published
- 2022
24. Strain and rupture of HIV-1 capsids during uncoating
- Author
-
Yu, Alvin, Lee, Elizabeth MY, Briggs, John AG, Ganser-Pornillos, Barbie K, Pornillos, Owen, and Voth, Gregory A
- Subjects
Infectious Diseases ,HIV/AIDS ,Aetiology ,2.2 Factors relating to the physical environment ,Infection ,Capsid ,Capsid Proteins ,Cell Line ,Electron Microscope Tomography ,HIV-1 ,Humans ,Molecular Dynamics Simulation ,Protein Conformation ,Virus Uncoating ,HIV capsid ,all-atom molecular dynamics ,  ,uncoating ,strain - Abstract
SignificanceThe mature capsids of HIV-1 are transiently stable complexes that self-assemble around the viral genome during maturation, and uncoat to release preintegration complexes that archive a double-stranded DNA copy of the virus in the host cell genome. However, a detailed view of how HIV cores rupture remains lacking. Here, we elucidate the physical properties involved in capsid rupture using a combination of large-scale all-atom molecular dynamics simulations and cryo-electron tomography. We find that intrinsic strain on the capsid forms highly correlated patterns along the capsid surface, along which cracks propagate. Capsid rigidity also increases with high strain. Our findings provide fundamental insight into viral capsid uncoating.
- Published
- 2022
25. Structures of Two Human Astrovirus Capsid/Neutralizing Antibody Complexes Reveal Distinct Epitopes and Inhibition of Virus Attachment to Cells
- Author
-
Ricemeyer, Lena, Aguilar-Hernández, Nayeli, López, Tomás, Espinosa, Rafaela, Lanning, Sarah, Mukherjee, Santanu, Cuellar, Carolina, López, Susana, Arias, Carlos F, and DuBois, Rebecca M
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Biotechnology ,Emerging Infectious Diseases ,Digestive Diseases ,Infectious Diseases ,Biodefense ,Immunization ,Prevention ,Vaccine Related ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,Amino Acid Sequence ,Antibodies ,Neutralizing ,Antibodies ,Viral ,Antibody Affinity ,Astroviridae Infections ,Capsid ,Capsid Proteins ,Epitopes ,Host-Pathogen Interactions ,Humans ,Mamastrovirus ,Models ,Molecular ,Molecular Conformation ,Protein Binding ,Structure-Activity Relationship ,Virus Attachment ,astrovirus ,capsid ,neutralizing antibodies ,protein structure-function ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
Human astrovirus is an important cause of viral gastroenteritis worldwide. Young children, the elderly, and the immunocompromised are especially at risk for contracting severe disease. However, no vaccines exist to combat human astrovirus infection. Evidence points to the importance of antibodies in protecting healthy adults from reinfection. To develop an effective subunit vaccine that broadly protects against diverse astrovirus serotypes, we must understand how neutralizing antibodies target the capsid surface at the molecular level. Here, we report the structures of the human astrovirus capsid spike domain bound to two neutralizing monoclonal antibodies. These antibodies bind two distinct conformational epitopes on the spike surface. We add to existing evidence that the human astrovirus capsid spike contains a receptor-binding domain and demonstrate that both antibodies neutralize human astrovirus by blocking virus attachment to host cells. We identify patches of conserved amino acids which overlap or border the antibody epitopes and may constitute a receptor-binding site. Our findings provide a basis for developing therapies to prevent and treat human astrovirus gastroenteritis. IMPORTANCE Human astroviruses infect nearly every person in the world during childhood and cause diarrhea, vomiting, and fever. Despite the prevalence of this virus, little is known about how antibodies block astrovirus infection. Here, we determined the crystal structures of the astrovirus capsid protein in complex with two virus-neutralizing antibodies. We show that the antibodies bind to two distinct sites on the capsid spike domain, however, both antibodies block virus attachment to human cells. Importantly, our findings support the use of the human astrovirus capsid spike as an antigen in a subunit-based vaccine to prevent astrovirus disease.
- Published
- 2022
26. Novel fold of rotavirus glycan-binding domain predicted by AlphaFold2 and determined by X-ray crystallography
- Author
-
Hu, Liya, Salmen, Wilhelm, Sankaran, Banumathi, Lasanajak, Yi, Smith, David F, Crawford, Sue E, Estes, Mary K, and Prasad, BV Venkataram
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Digestive Diseases ,Infection ,Capsid Proteins ,Child ,Crystallography ,X-Ray ,Galectins ,Humans ,Polysaccharides ,Rotavirus ,Biological sciences ,Biomedical and clinical sciences - Abstract
The VP8* domain of spike protein VP4 in group A and C rotaviruses, which cause epidemic gastroenteritis in children, exhibits a conserved galectin-like fold for recognizing glycans during cell entry. In group B rotavirus, which causes significant diarrheal outbreaks in adults, the VP8* domain (VP8*B) surprisingly lacks sequence similarity with VP8* of group A or group C rotavirus. Here, by using the recently developed AlphaFold2 for ab initio structure prediction and validating the predicted model by determining a 1.3-Å crystal structure, we show that VP8*B exhibits a novel fold distinct from the galectin fold. This fold with a β-sheet clasping an α-helix represents a new fold for glycan recognition based on glycan array screening, which shows that VP8*B recognizes glycans containing N-acetyllactosamine moiety. Although uncommon, our study illustrates how evolution can incorporate structurally distinct folds with similar functionality in a homologous protein within the same virus genus.
- Published
- 2022
27. Coat proteins of necroviruses target 14-3-3a to subvert MAPKKKα-mediated antiviral immunity in plants
- Author
-
Gao, Zongyu, Zhang, Dingliang, Wang, Xiaoling, Zhang, Xin, Wen, Zhiyan, Zhang, Qianshen, Li, Dawei, Dinesh-Kumar, Savithramma P, and Zhang, Yongliang
- Subjects
Infectious Diseases ,Infection ,14-3-3 Proteins ,Capsid Proteins ,Cell Death ,Gene Expression Regulation ,Plant ,Host-Pathogen Interactions ,Immune Evasion ,MAP Kinase Kinase Kinases ,MAP Kinase Signaling System ,Plant Diseases ,Plant Immunity ,Plant Leaves ,Protein Binding ,Tobacco ,Tombusviridae - Abstract
Mitogen-activated protein kinase (MAPK) cascades play an important role in innate immunity against various pathogens in plants and animals. However, we know very little about the importance of MAPK cascades in plant defense against viral pathogens. Here, we used a positive-strand RNA necrovirus, beet black scorch virus (BBSV), as a model to investigate the relationship between MAPK signaling and virus infection. Our findings showed that BBSV infection activates MAPK signaling, whereas viral coat protein (CP) counteracts MAPKKKα-mediated antiviral defense. CP does not directly target MAPKKKα, instead it competitively interferes with the binding of 14-3-3a to MAPKKKα in a dose-dependent manner. This results in the instability of MAPKKKα and subversion of MAPKKKα-mediated antiviral defense. Considering the conservation of 14-3-3-binding sites in the CPs of diverse plant viruses, we provide evidence that 14-3-3-MAPKKKα defense signaling module is a target of viral effectors in the ongoing arms race of defense and viral counter-defense.
- Published
- 2022
28. A defective viral genome strategy elicits broad protective immunity against respiratory viruses
- Author
-
Xiao, Yinghong, Lidsky, Peter V, Shirogane, Yuta, Aviner, Ranen, Wu, Chien-Ting, Li, Weiyi, Zheng, Weihao, Talbot, Dale, Catching, Adam, Doitsh, Gilad, Su, Weiheng, Gekko, Colby E, Nayak, Arabinda, Ernst, Joel D, Brodsky, Leonid, Brodsky, Elia, Rousseau, Elsa, Capponi, Sara, Bianco, Simone, Nakamura, Robert, Jackson, Peter K, Frydman, Judith, and Andino, Raul
- Subjects
Prevention ,Biodefense ,Pneumonia & Influenza ,Lung ,Immunization ,Vaccine Related ,Emerging Infectious Diseases ,Infectious Diseases ,Pneumonia ,Aetiology ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,Administration ,Intranasal ,Animals ,Antiviral Agents ,Broadly Neutralizing Antibodies ,COVID-19 ,Capsid Proteins ,Cell Line ,Defective Interfering Viruses ,Disease Models ,Animal ,Genome ,Viral ,Humans ,Influenza ,Human ,Interferons ,Male ,Mice ,Mice ,Inbred C57BL ,Poliovirus ,Respiratory Tract Infections ,SARS-CoV-2 ,Virus Replication ,RNA viruses ,antiviral ,broad-spectrum ,defective viral genomes ,innate immunity ,interferon ,respiratory infection ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
RNA viruses generate defective viral genomes (DVGs) that can interfere with replication of the parental wild-type virus. To examine their therapeutic potential, we created a DVG by deleting the capsid-coding region of poliovirus. Strikingly, intraperitoneal or intranasal administration of this genome, which we termed eTIP1, elicits an antiviral response, inhibits replication, and protects mice from several RNA viruses, including enteroviruses, influenza, and SARS-CoV-2. While eTIP1 replication following intranasal administration is limited to the nasal cavity, its antiviral action extends non-cell-autonomously to the lungs. eTIP1 broad-spectrum antiviral effects are mediated by both local and distal type I interferon responses. Importantly, while a single eTIP1 dose protects animals from SARS-CoV-2 infection, it also stimulates production of SARS-CoV-2 neutralizing antibodies that afford long-lasting protection from SARS-CoV-2 reinfection. Thus, eTIP1 is a safe and effective broad-spectrum antiviral generating short- and long-term protection against SARS-CoV-2 and other respiratory infections in animal models.
- Published
- 2021
29. Bluetongue virus capsid protein VP5 perforates membranes at low endosomal pH during viral entry
- Author
-
Xia, Xian, Wu, Weining, Cui, Yanxiang, Roy, Polly, and Zhou, Z Hong
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Emerging Infectious Diseases ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,1.1 Normal biological development and functioning ,Infection ,Animals ,Bluetongue ,Bluetongue virus ,Capsid Proteins ,Cryoelectron Microscopy ,Endosomes ,Hydrogen-Ion Concentration ,Models ,Molecular ,Sheep ,Sheep Diseases ,Virus Internalization ,Microbiology ,Medical Microbiology - Abstract
Bluetongue virus (BTV) is a non-enveloped virus and causes substantial morbidity and mortality in ruminants such as sheep. Fashioning a receptor-binding protein (VP2) and a membrane penetration protein (VP5) on the surface, BTV releases its genome-containing core (VP3 and VP7) into the host cell cytosol after perforation of the endosomal membrane. Unlike enveloped ones, the entry mechanisms of non-enveloped viruses into host cells remain poorly understood. Here we applied single-particle cryo-electron microscopy, cryo-electron tomography and structure-guided functional assays to characterize intermediate states of BTV cell entry in endosomes. Four structures of BTV at the resolution range of 3.4-3.9 Å show the different stages of structural rearrangement of capsid proteins on exposure to low pH, including conformational changes of VP5, stepwise detachment of VP2 and a small shift of VP7. In detail, sensing of the low-pH condition by the VP5 anchor domain triggers three major VP5 actions: projecting the hidden dagger domain, converting a surface loop to a protonated β-hairpin that anchors VP5 to the core and stepwise refolding of the unfurling domains into a six-helix stalk. Cryo-electron tomography structures of BTV interacting with liposomes show a length decrease of the VP5 stalk from 19.5 to 15.5 nm after its insertion into the membrane. Our structures, functional assays and structure-guided mutagenesis experiments combined indicate that this stalk, along with dagger domain and the WHXL motif, creates a single pore through the endosomal membrane that enables the viral core to enter the cytosol. Our study unveils the detailed mechanisms of BTV membrane penetration and showcases general methods to study cell entry of other non-enveloped viruses.
- Published
- 2021
30. Asymmetric reconstruction of mammalian reovirus reveals interactions among RNA, transcriptional factor µ2 and capsid proteins.
- Author
-
Pan, Muchen, Alvarez-Cabrera, Ana L, Kang, Joon S, Wang, Lihua, Fan, Chunhai, and Zhou, Z Hong
- Subjects
Cell Line ,Animals ,Macaca mulatta ,Orthoreovirus ,Nucleoside-Triphosphatase ,Transcription Factors ,Capsid Proteins ,RNA ,Double-Stranded ,RNA ,Messenger ,RNA ,Viral ,Cryoelectron Microscopy ,Virus Assembly ,Gene Expression Regulation ,Viral ,Allosteric Regulation ,Genome ,Viral ,Transcriptional Activation ,Protein Multimerization ,RNA-Dependent RNA Polymerase ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance - Abstract
Mammalian reovirus (MRV) is the prototypical member of genus Orthoreovirus of family Reoviridae. However, lacking high-resolution structures of its RNA polymerase cofactor μ2 and infectious particle, limits understanding of molecular interactions among proteins and RNA, and their contributions to virion assembly and RNA transcription. Here, we report the 3.3 Å-resolution asymmetric reconstruction of transcribing MRV and in situ atomic models of its capsid proteins, the asymmetrically attached RNA-dependent RNA polymerase (RdRp) λ3, and RdRp-bound nucleoside triphosphatase μ2 with a unique RNA-binding domain. We reveal molecular interactions among virion proteins and genomic and messenger RNA. Polymerase complexes in three Spinoreovirinae subfamily members are organized with different pseudo-D3d symmetries to engage their highly diversified genomes. The above interactions and those between symmetry-mismatched receptor-binding σ1 trimers and RNA-capping λ2 pentamers balance competing needs of capsid assembly, external protein removal, and allosteric triggering of endogenous RNA transcription, before, during and after infection, respectively.
- Published
- 2021
31. A Tail Fiber Protein and a Receptor-Binding Protein Mediate ICP2 Bacteriophage Interactions with Vibrio cholerae OmpU
- Author
-
Lim, Andrea NW, Yen, Minmin, Seed, Kimberley D, Lazinski, David W, and Camilli, Andrew
- Subjects
Foodborne Illness ,Infectious Diseases ,Genetics ,Emerging Infectious Diseases ,Biodefense ,Vaccine Related ,Digestive Diseases ,Prevention ,Aetiology ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,Adhesins ,Bacterial ,Alleles ,Animals ,Antigens ,Bacterial ,Bacteriophages ,Capsid Proteins ,Cholera ,Host Microbial Interactions ,Host Specificity ,Humans ,Inositol Phosphates ,Models ,Animal ,Mutation ,Mutation ,Missense ,Phenotype ,Porins ,Rabbits ,Vibrio cholerae ,Viral Tail Proteins ,OmpU ,tail fiber protein ,arms race ,bacteriophages ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Microbiology - Abstract
ICP2 is a virulent bacteriophage (phage) that preys on Vibrio cholerae. ICP2 was first isolated from cholera patient stool samples. Some of these stools also contained ICP2-resistant isogenic V. cholerae strains harboring missense mutations in the trimeric outer membrane porin protein OmpU, identifying it as the ICP2 receptor. In this study, we identify the ICP2 proteins that mediate interactions with OmpU by selecting for ICP2 host range mutants within infant rabbits infected with a mixture of wild-type and OmpU mutant strains. ICP2 host range mutants that can now infect OmpU mutant strains have missense mutations in the putative tail fiber gene gp25 and the putative adhesin gene gp23. Using site-specific mutagenesis, we show that single or double mutations in gp25 are sufficient to generate the host range mutant phenotype. However, at least one additional mutation in gp23 is required for robust plaque formation on specific OmpU mutants. Mutations in gp23 alone were insufficient to produce a host range mutant phenotype. All ICP2 host range mutants retained the ability to form plaques on wild-type V. cholerae cells. The strength of binding of host range mutants to V. cholerae correlated with plaque morphology, indicating that the selected mutations in gp25 and gp23 restore molecular interactions with the receptor. We propose that ICP2 host range mutants evolve by a two-step process. First, gp25 mutations are selected for their broad host range, albeit accompanied by low-level phage adsorption. Subsequent selection occurs for gp23 mutations that further increase productive binding to specific OmpU alleles, allowing for near-wild-type efficiencies of adsorption and subsequent phage multiplication. IMPORTANCE Concern over multidrug-resistant bacterial pathogens, including Vibrio cholerae, has led to renewed interest in phage biology and the potential for phage therapy. ICP2 is a genetically unique virulent phage isolated from cholera patient stool samples. It is also one of three phages in a prophylactic cocktail that have been shown to be effective in animal models of infection and the only one of the three that requires a protein receptor (OmpU). This study identifies an ICP2 tail fiber and a receptor binding protein and examines how ICP2 responds to the selective pressures of phage-resistant OmpU mutants. We found that this particular coevolutionary arms race presents fitness costs to both ICP2 and V. cholerae.
- Published
- 2021
32. A phage satellite tunes inducing phage gene expression using a domesticated endonuclease to balance inhibition and virion hijacking
- Author
-
Netter, Zoe, Boyd, Caroline M, Silvas, Tania V, and Seed, Kimberley D
- Subjects
Genetics ,Infectious Diseases ,Infection ,Bacterial Proteins ,Bacteriophages ,Binding Sites ,CRISPR-Cas Systems ,Capsid Proteins ,DNA ,Satellite ,DNA-Binding Proteins ,Endonucleases ,Gene Expression Regulation ,Viral ,Interspersed Repetitive Sequences ,Operon ,Protein Domains ,Transduction ,Genetic ,Vibrio cholerae ,Virion ,Environmental Sciences ,Biological Sciences ,Information and Computing Sciences ,Developmental Biology - Abstract
Bacteria persist under constant threat of predation by bacterial viruses (phages). Bacteria-phage conflicts result in evolutionary arms races often driven by mobile genetic elements (MGEs). One such MGE, a phage satellite in Vibrio cholerae called PLE, provides specific and robust defense against a pervasive lytic phage, ICP1. The interplay between PLE and ICP1 has revealed strategies for molecular parasitism allowing PLE to hijack ICP1 processes in order to mobilize. Here, we describe the mechanism of PLE-mediated transcriptional manipulation of ICP1 structural gene transcription. PLE encodes a novel DNA binding protein, CapR, that represses ICP1's capsid morphogenesis operon. Although CapR is sufficient for the degree of capsid repression achieved by PLE, its activity does not hinder the ICP1 lifecycle. We explore the consequences of repression of this operon, demonstrating that more stringent repression achieved through CRISPRi restricts both ICP1 and PLE. We also discover that PLE transduces in modified ICP1-like particles. Examination of CapR homologs led to the identification of a suite of ICP1-encoded homing endonucleases, providing a putative origin for the satellite-encoded repressor. This work unveils a facet of the delicate balance of satellite-mediated inhibition aimed at blocking phage production while successfully mobilizing in a phage-derived particle.
- Published
- 2021
33. Atomic Structure of the Trichomonas vaginalis Double-Stranded RNA Virus 2
- Author
-
Stevens, Alexander, Muratore, Katherine, Cui, Yanxiang, Johnson, Patricia J, and Zhou, Z Hong
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Genetics ,Sexually Transmitted Infections ,2.2 Factors relating to the physical environment ,1.1 Normal biological development and functioning ,Infection ,Good Health and Well Being ,Capsid ,Capsid Proteins ,Cryoelectron Microscopy ,Genome ,Viral ,Protein Conformation ,alpha-Helical ,RNA Viruses ,RNA ,Double-Stranded ,RNA ,Viral ,Totiviridae ,Trichomonas vaginalis ,cryoEM ,subparticle reconstruction ,double-stranded RNA virus ,Microbiology ,Biochemistry and cell biology ,Medical microbiology - Abstract
Trichomonas vaginalis, the causative pathogen for the most common nonviral sexually transmitted infection worldwide, is itself frequently infected with one or more of the four types of small double-stranded RNA (dsRNA) Trichomonas vaginalis viruses (TVV1 to 4, genus Trichomonasvirus, family Totiviridae). Each TVV encloses a nonsegmented genome within a single-layered capsid and replicates entirely intracellularly, like many dsRNA viruses, and unlike those in the Reoviridae family. Here, we have determined the structure of TVV2 by cryo-electron microscopy (cryoEM) at 3.6 Å resolution and derived an atomic model of its capsid. TVV2 has an icosahedral, T = 2*, capsid comprised of 60 copies of the icosahedral asymmetric unit (a dimer of the two capsid shell protein [CSP] conformers, CSP-A and CSP-B), typical of icosahedral dsRNA virus capsids. However, unlike the robust CSP-interlocking interactions such as the use of auxiliary "clamping" proteins among Reoviridae, only lateral CSP interactions are observed in TVV2, consistent with an assembly strategy optimized for TVVs' intracellular-only replication cycles within their protozoan host. The atomic model reveals both a mostly negatively charged capsid interior, which is conducive to movement of the loosely packed genome, and channels at the 5-fold vertices, which we suggest as routes of mRNA release during transcription. Structural comparison of TVV2 to the Saccharomyces cerevisiae L-A virus reveals a conserved helix-rich fold within the CSP and putative guanylyltransferase domain along the capsid exterior, suggesting conserved mRNA maintenance strategies among Totiviridae This first atomic structure of a TVV provides a framework to guide future biochemical investigations into the interplay between Trichomonas vaginalis and its viruses.IMPORTANCETrichomonas vaginalis viruses (TVVs) are double-stranded RNA (dsRNA) viruses that cohabitate in Trichomonas vaginalis, the causative pathogen of trichomoniasis, the most common nonviral sexually transmitted disease worldwide. Featuring an unsegmented dsRNA genome encoding a single capsid shell protein (CSP), TVVs contrast with multisegmented dsRNA viruses, such as the diarrhea-causing rotavirus, whose larger genome is split into 10 dsRNA segments encoding 5 unique capsid proteins. To determine how TVVs incorporate the requisite functionalities for viral replication into their limited proteome, we derived the atomic model of TVV2, a first for TVVs. Our results reveal the intersubunit interactions driving CSP association for capsid assembly and the properties that govern organization and maintenance of the viral genome. Structural comparison between TVV2 capsids and those of distantly related dsRNA viruses indicates conserved strategies of nascent RNA release and a putative viral guanylyltransferase domain implicated in the cytoplasmic maintenance of viral messenger and genomic RNA.
- Published
- 2021
34. Cryo-EM structures reveal the molecular basis of receptor-initiated coxsackievirus uncoating
- Author
-
Xu, Longfa, Zheng, Qingbing, Zhu, Rui, Yin, Zhichao, Yu, Hai, Lin, Yu, Wu, Yuanyuan, He, Maozhou, Huang, Yang, Jiang, Yichao, Sun, Hui, Zha, Zhenghui, Yang, Hongwei, Huang, Qiongzi, Zhang, Dongqing, Chen, Zhenqin, Ye, Xiangzhong, Han, Jinle, Yang, Lisheng, Liu, Che, Que, Yuqiong, Fang, Mujin, Gu, Ying, Zhang, Jun, Luo, Wenxin, Zhou, Z Hong, Li, Shaowei, Cheng, Tong, and Xia, Ningshao
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Infectious Diseases ,Biotechnology ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Infection ,Animals ,Antibodies ,Neutralizing ,Capsid ,Capsid Proteins ,Cryoelectron Microscopy ,Enterovirus ,Enterovirus B ,Human ,Enterovirus Infections ,Female ,Mice ,Mice ,Inbred BALB C ,Models ,Molecular ,Protein Interaction Domains and Motifs ,Receptors ,Virus ,Virion ,Virus Uncoating ,coxsackievirus ,coxsackievirus and adenovirus receptor ,cryoEM structures ,neutralizing antibody ,virus uncoating ,Microbiology ,Medical Microbiology ,Biochemistry and cell biology ,Medical microbiology - Abstract
Enterovirus uncoating receptors bind at the surface depression ("canyon") that encircles each capsid vertex causing the release of a host-derived lipid called "pocket factor" that is buried in a hydrophobic pocket formed by the major viral capsid protein, VP1. Coxsackievirus and adenovirus receptor (CAR) is a universal uncoating receptor of group B coxsackieviruses (CVB). Here, we present five high-resolution cryoEM structures of CVB representing different stages of virus infection. Structural comparisons show that the CAR penetrates deeper into the canyon than other uncoating receptors, leading to a cascade of events: collapse of the VP1 hydrophobic pocket, high-efficiency release of the pocket factor and viral uncoating and genome release under neutral pH, as compared with low pH. Furthermore, we identified a potent therapeutic antibody that can neutralize viral infection by interfering with virion-CAR interactions, destabilizing the capsid and inducing virion disruption. Together, these results define the structural basis of CVB cell entry and antibody neutralization.
- Published
- 2021
35. Plant-expressed virus-like particles reveal the intricate maturation process of a eukaryotic virus
- Author
-
Castells-Graells, Roger, Ribeiro, Jonas RS, Domitrovic, Tatiana, Hesketh, Emma L, Scarff, Charlotte A, Johnson, John E, Ranson, Neil A, Lawson, David M, and Lomonossoff, George P
- Subjects
Plant Biology ,Biological Sciences ,Capsid Proteins ,Cryoelectron Microscopy ,Eukaryota ,Hydrogen-Ion Concentration ,Models ,Molecular ,Plant Leaves ,Protein Structure ,Quaternary ,RNA Viruses ,Tobacco ,Virion ,Virus Assembly ,Biological sciences ,Biomedical and clinical sciences - Abstract
Many virus capsids undergo exquisitely choreographed maturation processes in their host cells to produce infectious virions, and these remain poorly understood. As a tool for studying virus maturation, we transiently expressed the capsid protein of the insect virus Nudaurelia capensis omega virus (NωV) in Nicotiana benthamiana and were able to purify both immature procapsids and mature capsids from infiltrated leaves by varying the expression time. Cryo-EM analysis of the plant-produced procapsids and mature capsids to 6.6 Å and 2.7 Å resolution, respectively, reveals that in addition to large scale rigid body motions, internal regions of the subunits are extensively remodelled during maturation, creating the active site required for autocatalytic cleavage and infectivity. The mature particles are biologically active in terms of their ability to lyse membranes and have a structure that is essentially identical to authentic virus. The ability to faithfully recapitulate and visualize a complex maturation process in plants, including the autocatalytic cleavage of the capsid protein, has revealed a ~30 Å translation-rotation of the subunits during maturation as well as conformational rearrangements in the N and C-terminal helical regions of each subunit.
- Published
- 2021
36. Asymmetrizing an icosahedral virus capsid by hierarchical assembly of subunits with designed asymmetry
- Author
-
Zhao, Zhongchao, Wang, Joseph Che-Yen, Zhang, Mi, Lyktey, Nicholas A, Jarrold, Martin F, Jacobson, Stephen C, and Zlotnick, Adam
- Subjects
Biological Sciences ,Chemical Sciences ,Physical Chemistry ,Infectious Diseases ,Bioengineering ,Infection ,Good Health and Well Being ,Capsid ,Capsid Proteins ,Cryoelectron Microscopy ,Hepatitis B virus ,Models ,Molecular ,Protein Multimerization ,Recombinant Proteins ,Virus Assembly - Abstract
Symmetrical protein complexes are ubiquitous in biology. Many have been re-engineered for chemical and medical applications. Viral capsids and their assembly are frequent platforms for these investigations. A means to create asymmetric capsids may expand applications. Here, starting with homodimeric Hepatitis B Virus capsid protein, we develop a heterodimer, design a hierarchical assembly pathway, and produce asymmetric capsids. In the heterodimer, the two halves have different growth potentials and assemble into hexamers. These preformed hexamers can nucleate co-assembly with other dimers, leading to Janus-like capsids with a small discrete hexamer patch. We can remove the patch specifically and observe asymmetric holey capsids by cryo-EM reconstruction. The resulting hole in the surface can be refilled with fluorescently labeled dimers to regenerate an intact capsid. In this study, we show how an asymmetric subunit can be used to generate an asymmetric particle, creating the potential for a capsid with different surface chemistries.
- Published
- 2021
37. In vivo pair correlation microscopy reveals dengue virus capsid protein nucleocytoplasmic bidirectional movement in mammalian infected cells
- Author
-
Sallaberry, Ignacio, Luszczak, Alexis, Philipp, Natalia, Navarro, Guadalupe S Costa, Gabriel, Manuela V, Gratton, Enrico, Gamarnik, Andrea V, and Estrada, Laura C
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Biodefense ,Prevention ,Vaccine Related ,Infectious Diseases ,Rare Diseases ,Emerging Infectious Diseases ,Vector-Borne Diseases ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Good Health and Well Being ,Active Transport ,Cell Nucleus ,Animals ,Capsid Proteins ,Cell Line ,Cricetinae ,Dengue ,Dengue Virus - Abstract
Flaviviruses are major human disease-causing pathogens, including dengue virus (DENV), Zika virus, yellow fever virus and others. DENV infects hundreds of millions of people per year around the world, causing a tremendous social and economic burden. DENV capsid (C) protein plays an essential role during genome encapsidation and viral particle formation. It has been previously shown that DENV C enters the nucleus in infected cells. However, whether DENV C protein exhibits nuclear export remains unclear. By spatially cross-correlating different regions of the cell, we investigated DENV C movement across the nuclear envelope during the infection cycle. We observed that transport takes place in both directions and with similar translocation times (in the ms time scale) suggesting a bidirectional movement of both C protein import and export.Furthermore, from the pair cross-correlation functions in cytoplasmic or nuclear regions we found two populations of C molecules in each compartment with fast and slow mobilities. While in the cytoplasm the correlation times were in the 2-6 and 40-110 ms range for the fast and slow mobility populations respectively, in the cell nucleus they were 1-10 and 25-140 ms range, respectively. The fast mobility of DENV C in cytoplasmic and nuclear regions agreed with the diffusion coefficients from Brownian motion previously reported from correlation analysis. These studies provide the first evidence of DENV C shuttling from and to the nucleus in infected cells, opening new venues for antiviral interventions.
- Published
- 2021
38. Protein Disulfide Isomerase A4 Is Involved in Genome Uncoating during Human Astrovirus Cell Entry
- Author
-
Aguilar-Hernández, Nayeli, Meyer, Lena, López, Susana, DuBois, Rebecca M, and Arias, Carlos F
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biotechnology ,Digestive Diseases ,Emerging Infectious Diseases ,Infectious Diseases ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Astroviridae Infections ,Capsid Proteins ,Cell Line ,Cells ,Cultured ,Host-Pathogen Interactions ,Humans ,Mamastrovirus ,Protein Binding ,Protein Disulfide-Isomerases ,Virus Internalization ,Virus Uncoating ,astrovirus ,protein disulfide isomerase ,virus entry ,Microbiology - Abstract
Although human astroviruses (HAstVs) are important agents of gastroenteritis in young children, the studies aimed at characterizing their biology have been limited, in particular regarding their cell entry process. It has been shown that HAstV serotype 8 enters human cells by a classical clathrin-mediated endocytosis pathway; however, the cell receptor or other cell entry factors that may be relevant for an efficient viral infection are unknown. In this work we used a far-Western blotting approach to identify cellular proteins that interact with the recombinant capsid spike proteins of HAstV serotypes 1, 2, and 8, synthesized in Escherichia coli. We identified the 72 kDa protein disulfide isomerase A4 (PDIA4) as a binding partner for HAstV-1 and -8 spikes, but not for the HAstV-2 spike. In agreement with this observation, the PDI inhibitor 16F16 strongly blocked infection by HAstV serotypes 1 and 8, but not serotype 2. RNA interference of PDIA4 expression selectively blocked HAstV-8 infectivity. We also showed that the PDI activity does not affect virus binding or internalization but is required for uncoating of the viral genome.
- Published
- 2021
39. Human Astrovirus 1–8 Seroprevalence Evaluation in a United States Adult Population
- Author
-
Meyer, Lena, Delgado-Cunningham, Kevin, Lorig-Roach, Nicholas, Ford, Jordan, and DuBois, Rebecca M
- Subjects
Microbiology ,Biological Sciences ,Health Disparities ,Immunization ,Digestive Diseases ,Infectious Diseases ,Clinical Research ,Infection ,Adult ,Age Factors ,Antibodies ,Neutralizing ,Astroviridae Infections ,Capsid ,Capsid Proteins ,Humans ,Immunosorbent Techniques ,Mamastrovirus ,Population Surveillance ,Seroepidemiologic Studies ,Serogroup ,United States ,human astrovirus ,seroprevalence ,biolayer interferometry immunosorbent assay - Abstract
Human astroviruses are an important cause of viral gastroenteritis globally, yet few studies have investigated the serostatus of adults to establish rates of previous infection. Here, we applied biolayer interferometry immunosorbent assay (BLI-ISA), a recently developed serosurveillance technique, to measure the presence of blood plasma IgG antibodies directed towards the human astrovirus capsid spikes from serotypes 1-8 in a cross-sectional sample of a United States adult population. The seroprevalence rates of IgG antibodies were 73% for human astrovirus serotype 1, 62% for serotype 3, 52% for serotype 4, 29% for serotype 5, 27% for serotype 8, 22% for serotype 2, 8% for serotype 6, and 8% for serotype 7. Notably, seroprevalence rates for capsid spike antigens correlate with neutralizing antibody rates determined previously. This work is the first seroprevalence study evaluating all eight classical human astrovirus serotypes.
- Published
- 2021
40. Protein Disulfide Isomerase A4 Is Involved in Genome Uncoating during Human Astrovirus Cell Entry.
- Author
-
Aguilar-Hernández, Nayeli, Meyer, Lena, López, Susana, DuBois, Rebecca M, and Arias, Carlos F
- Subjects
Cells ,Cultured ,Cell Line ,Humans ,Astroviridae Infections ,Capsid Proteins ,Protein Binding ,Mamastrovirus ,Virus Internalization ,Host-Pathogen Interactions ,Protein Disulfide-Isomerases ,Virus Uncoating ,astrovirus ,protein disulfide isomerase ,virus entry ,Cells ,Cultured ,Microbiology - Abstract
Although human astroviruses (HAstVs) are important agents of gastroenteritis in young children, the studies aimed at characterizing their biology have been limited, in particular regarding their cell entry process. It has been shown that HAstV serotype 8 enters human cells by a classical clathrin-mediated endocytosis pathway; however, the cell receptor or other cell entry factors that may be relevant for an efficient viral infection are unknown. In this work we used a far-Western blotting approach to identify cellular proteins that interact with the recombinant capsid spike proteins of HAstV serotypes 1, 2, and 8, synthesized in Escherichia coli. We identified the 72 kDa protein disulfide isomerase A4 (PDIA4) as a binding partner for HAstV-1 and -8 spikes, but not for the HAstV-2 spike. In agreement with this observation, the PDI inhibitor 16F16 strongly blocked infection by HAstV serotypes 1 and 8, but not serotype 2. RNA interference of PDIA4 expression selectively blocked HAstV-8 infectivity. We also showed that the PDI activity does not affect virus binding or internalization but is required for uncoating of the viral genome.
- Published
- 2020
41. The Integrity of the Intradimer Interface of the Hepatitis B Virus Capsid Protein Dimer Regulates Capsid Self-Assembly
- Author
-
Zhao, Zhongchao, Wang, Joseph Che-Yen, Segura, Carolina Pérez, Hadden-Perilla, Jodi A, and Zlotnick, Adam
- Subjects
Biological Sciences ,Macromolecular and Materials Chemistry ,Chemical Sciences ,Liver Disease ,Hepatitis ,Digestive Diseases ,Infectious Diseases ,Bioengineering ,HIV/AIDS ,Infection ,Good Health and Well Being ,Capsid ,Capsid Proteins ,Dimerization ,Hepatitis B virus ,Molecular Dynamics Simulation ,Protein Conformation ,Reproducibility of Results ,Organic Chemistry ,Biological sciences ,Chemical sciences - Abstract
During the hepatitis B virus lifecycle, 120 copies of homodimeric capsid protein assemble around a copy of reverse transcriptase and viral RNA and go on to produce an infectious virion. Assembly needs to be tightly regulated by protein conformational change to ensure symmetry, fidelity, and reproducibility. Here, we show that structures at the intradimer interface regulate conformational changes at the distal interdimer interface and so regulate assembly. A pair of interacting charged residues, D78 from each monomer, conspicuously located at the top of a four-helix bundle that forms the intradimer interface, were mutated to serine to disrupt communication between the two monomers. The mutation slowed assembly and destabilized the dimer to thermal and chemical denaturation. Mutant dimers showed evidence of transient partial unfolding based on the appearance of new proteolytically sensitive sites. Though the mutant dimer was less stable, the resulting capsids were as stable as the wildtype, based on assembly and thermal denaturation studies. Cryo-EM image reconstructions of capsid indicated that the subunits adopted an "open" state more usually associated with a free dimer and that the spike tips were either disordered or highly flexible. Molecular dynamics simulations provide mechanistic explanations for these results, suggesting that D78 stabilizes helix 4a, which forms part of the intradimer interface, by capping its N-terminus and hydrogen-bonding to nearby residues, whereas the D78S mutation disrupts these interactions, leading to partial unwinding of helix 4a. This in turn weakens the connection from helix 4 and the intradimer interface to helix 5, which forms the interdimer interface.
- Published
- 2020
42. Near-atomic cryo-electron microscopy structures of varicella-zoster virus capsids
- Author
-
Wang, Wei, Zheng, Qingbing, Pan, Dequan, Yu, Hai, Fu, Wenkun, Liu, Jian, He, Maozhou, Zhu, Rui, Cai, Yuze, Huang, Yang, Zha, Zhenghui, Chen, Zhenqin, Ye, Xiangzhong, Han, Jinle, Que, Yuqiong, Wu, Ting, Zhang, Jun, Li, Shaowei, Zhu, Hua, Zhou, Z Hong, Cheng, Tong, and Xia, Ningshao
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Vaccine Related ,Infectious Diseases ,Immunization ,Biotechnology ,Infection ,Capsid ,Capsid Proteins ,Cryoelectron Microscopy ,Herpesvirus 3 ,Human ,Humans ,Models ,Molecular ,Protein Domains ,Varicella Zoster Virus Infection ,Virion ,Microbiology ,Medical Microbiology - Abstract
Varicella-zoster virus (VZV) is a medically important human herpesvirus that causes chickenpox and shingles, but its cell-associated nature has hindered structure studies. Here we report the cryo-electron microscopy structures of purified VZV A-capsid and C-capsid, as well as of the DNA-containing capsid inside the virion. Atomic models derived from these structures show that, despite enclosing a genome that is substantially smaller than those of other human herpesviruses, VZV has a similarly sized capsid, consisting of 955 major capsid protein (MCP), 900 small capsid protein (SCP), 640 triplex dimer (Tri2) and 320 triplex monomer (Tri1) subunits. The VZV capsid has high thermal stability, although with relatively fewer intra- and inter-capsid protein interactions and less stably associated tegument proteins compared with other human herpesviruses. Analysis with antibodies targeting the N and C termini of the VZV SCP indicates that the hexon-capping SCP-the largest among human herpesviruses-uses its N-terminal half to bridge hexon MCP subunits and possesses a C-terminal flexible half emanating from the inner rim of the upper hexon channel into the tegument layer. Correlation of these structural features and functional observations provide insights into VZV assembly and pathogenesis and should help efforts to engineer gene delivery and anticancer vectors based on the currently available VZV vaccine.
- Published
- 2020
43. Engineering a Virus-like Particle to Display Peptide Insertions Using an Apparent Fitness Landscape
- Author
-
Robinson, Stephanie A, Hartman, Emily C, Ikwuagwu, Bon C, Francis, Matthew B, and Tullman-Ercek, Danielle
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Bioengineering ,Biotechnology ,Generic health relevance ,Amino Acid Sequence ,Capsid ,Capsid Proteins ,Peptides ,Vaccines ,Virus-Like Particle ,Chemical Sciences ,Engineering ,Polymers ,Biological sciences ,Chemical sciences - Abstract
Peptide insertions in the primary sequence of proteins expand functionality by introducing new binding sequences, chemical handles, or membrane disrupting motifs. With these properties, proteins can be engineered as scaffolds for vaccines or targeted drug delivery vehicles. Virus-like particles (VLPs) are promising platforms for these applications since they are genetically simple, mimic viral structure for cell uptake, and can deliver multiple copies of a therapeutic agent to a given cell. Peptide insertions in the coat protein of VLPs can increase VLP uptake in cells by increasing cell binding, but it is difficult to predict how an insertion affects monomer folding and higher order assembly. To this end, we have engineered the MS2 VLP using a high-throughput technique, called Systematic Mutagenesis and Assembled Particle Selection (SyMAPS). In this work, we applied SyMAPS to investigate a highly mutable loop in the MS2 coat protein to display 9,261 non-native tripeptide insertions. This library generates a discrete map of three amino acid insertions permitted at this location, validates the FG loop as a valuable position for peptide insertion, and illuminates how properties such as charge, flexibility, and hydrogen bonding can interact to preserve or disrupt capsid assembly. Taken together, the results highlight the potential to engineer VLPs in a systematic manner, paving the way to exploring the applications of peptide insertions in biomedically relevant settings.
- Published
- 2020
44. Structures of capsid and capsid-associated tegument complex inside the Epstein–Barr virus
- Author
-
Liu, Wei, Cui, Yanxiang, Wang, Caiyan, Li, Zihang, Gong, Danyang, Dai, Xinghong, Bi, Guo-Qiang, Sun, Ren, and Zhou, Z Hong
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Lymphatic Research ,Cancer ,Hematology ,Lymphoma ,Rare Diseases ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Infection ,Amino Acid Sequence ,Capsid ,Capsid Proteins ,Cryoelectron Microscopy ,Epstein-Barr Virus Infections ,Herpesvirus 4 ,Human ,Humans ,Imaging ,Three-Dimensional ,Models ,Molecular ,Protein Subunits ,Structure-Activity Relationship ,Virion ,Virus Assembly ,Microbiology ,Medical Microbiology - Abstract
As the first discovered human cancer virus, Epstein-Barr virus (EBV) causes Burkitt's lymphoma and nasopharyngeal carcinoma. Isolating virions for determining high-resolution structures has been hindered by latency-a hallmark of EBV infection-and atomic structures are thus available only for recombinantly expressed EBV proteins. In the present study, by symmetry relaxation and subparticle reconstruction, we have determined near-atomic-resolution structures of the EBV capsid with an asymmetrically attached DNA-translocating portal and capsid-associated tegument complexes from cryogenic electron microscopy images of just 2,048 EBV virions obtained by chemical induction. The resulting atomic models reveal structural plasticity among the 20 conformers of the major capsid protein, 2 conformers of the small capsid protein (SCP), 4 conformers of the triplex monomer proteins and 2 conformers of the triplex dimer proteins. Plasticity reaches the greatest level at the capsid-tegument interfaces involving SCP and capsid-associated tegument complexes (CATC): SCPs crown pentons/hexons and mediate tegument protein binding, and CATCs bind and rotate all five periportal triplexes, but notably only about one peri-penton triplex. These results offer insights into the EBV capsid assembly and a mechanism for recruiting cell-regulating factors into the tegument compartment as 'cargoes', and should inform future anti-EBV strategies.
- Published
- 2020
45. Mapping Attenuation Determinants in Enterovirus-D68.
- Author
-
Yeh, Ming Te, Capponi, Sara, Catching, Adam, Bianco, Simone, and Andino, Raul
- Subjects
Brain ,Spinal Cord ,Cell Line ,Cell Line ,Tumor ,Animals ,Mice ,Inbred C57BL ,Mice ,Knockout ,Humans ,Enterovirus D ,Human ,Enterovirus Infections ,Disease Models ,Animal ,Capsid Proteins ,5' Untranslated Regions ,Amino Acid Substitution ,Virus Replication ,Virulence ,Genes ,Viral ,Models ,Molecular ,Receptor ,Interferon alpha-beta ,Molecular Dynamics Simulation ,VP3 ,enterovirus ,enterovirus-D68 ,infectious clones ,mouse model ,paralysis ,virulence determinant ,Microbiology - Abstract
Enterovirus (EV)-D68 has been associated with epidemics in the United Sates in 2014, 2016 and 2018. This study aims to identify potential viral virulence determinants. We found that neonatal type I interferon receptor knockout mice are susceptible to EV-D68 infection via intraperitoneal inoculation and were able to recapitulate the paralysis process observed in human disease. Among the EV-D68 strains tested, strain US/MO-14-18949 caused no observable disease in this mouse model, whereas the other strains caused paralysis and death. Sequence analysis revealed several conserved genetic changes among these virus strains: nucleotide positions 107 and 648 in the 5'-untranslated region (UTR); amino acid position 88 in VP3; 1, 148, 282 and 283 in VP1; 22 in 2A; 47 in 3A. A series of chimeric and point-mutated infectious clones were constructed to identify viral elements responsible for the distinct virulence. A single amino acid change from isoleucine to valine at position 88 in VP3 attenuated neurovirulence by reducing virus replication in the brain and spinal cord of infected mice.
- Published
- 2020
46. Structural basis for capsid recruitment and coat formation during HSV-1 nuclear egress.
- Author
-
Draganova, Elizabeth B, Zhang, Jiayan, Zhou, Z Hong, and Heldwein, Ekaterina E
- Subjects
Escherichia coli ,Herpesvirus 1 ,Human ,Capsid Proteins ,Microscopy ,Confocal ,Cryoelectron Microscopy ,Cloning ,Molecular ,Virus Replication ,Gene Expression Regulation ,Viral ,Active Transport ,Cell Nucleus ,Protein Transport ,capsid budding ,herpes simplex virus type 1 ,herpesvirus ,infectious disease ,membrane budding ,microbiology ,nuclear egress ,nuclear egress complex ,virus ,Rare Diseases ,2.2 Factors relating to the physical environment ,Infection ,Biochemistry and Cell Biology - Abstract
During herpesvirus infection, egress of nascent viral capsids from the nucleus is mediated by the viral nuclear egress complex (NEC). NEC deforms the inner nuclear membrane (INM) around the capsid by forming a hexagonal array. However, how the NEC coat interacts with the capsid and how curved coats are generated to enable budding is yet unclear. Here, by structure-guided truncations, confocal microscopy, and cryoelectron tomography, we show that binding of the capsid protein UL25 promotes the formation of NEC pentagons rather than hexagons. We hypothesize that during nuclear budding, binding of UL25 situated at the pentagonal capsid vertices to the NEC at the INM promotes formation of NEC pentagons that would anchor the NEC coat to the capsid. Incorporation of NEC pentagons at the points of contact with the vertices would also promote assembly of the curved hexagonal NEC coat around the capsid, leading to productive egress of UL25-decorated capsids.
- Published
- 2020
47. Genome organization and interaction with capsid protein in a multipartite RNA virus
- Author
-
Beren, Christian, Cui, Yanxiang, Chakravarty, Antara, Yang, Xue, Rao, ALN, Knobler, Charles M, Zhou, Z Hong, and Gelbart, William M
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Infectious Diseases ,Genetics ,Infection ,Bacteriophages ,Bromovirus ,Capsid Proteins ,Genome ,Viral ,RNA ,Viral ,cryoelectron microscopy ,virus ,single-stranded RNA - Abstract
We report the asymmetric reconstruction of the single-stranded RNA (ssRNA) content in one of the three otherwise identical virions of a multipartite RNA virus, brome mosaic virus (BMV). We exploit a sample consisting exclusively of particles with the same RNA content-specifically, RNAs 3 and 4-assembled in planta by agrobacterium-mediated transient expression. We find that the interior of the particle is nearly empty, with most of the RNA genome situated at the capsid shell. However, this density is disordered in the sense that the RNA is not associated with any particular structure but rather, with an ensemble of secondary/tertiary structures that interact with the capsid protein. Our results illustrate a fundamental difference between the ssRNA organization in the multipartite BMV viral capsid and the monopartite bacteriophages MS2 and Qβ for which a dominant RNA conformation is found inside the assembled viral capsids, with RNA density conserved even at the center of the particle. This can be understood in the context of the differing demands on their respective lifecycles: BMV must package separately each of several different RNA molecules and has been shown to replicate and package them in isolated, membrane-bound, cytoplasmic complexes, whereas the bacteriophages exploit sequence-specific "packaging signals" throughout the viral RNA to package their monopartite genomes.
- Published
- 2020
48. 2.7 Å cryo-EM structure of rotavirus core protein VP3, a unique capping machine with a helicase activity
- Author
-
Kumar, Dilip, Yu, Xinzhe, Crawford, Sue E, Moreno, Rodolfo, Jakana, Joanita, Sankaran, Banumathi, Anish, Ramakrishnan, Kaundal, Soni, Hu, Liya, Estes, Mary K, Wang, Zhao, and Prasad, BV Venkataram
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Infectious Diseases ,Digestive Diseases ,Foodborne Illness ,Capsid Proteins ,Cryoelectron Microscopy ,Genome ,Viral ,Nucleotidyltransferases ,RNA ,Viral ,Rotavirus ,Virion - Abstract
In many viruses, including rotavirus (RV), the major pathogen of infantile gastroenteritis, capping of viral messenger RNAs is a pivotal step for efficient translation of the viral genome. In RV, VP3 caps the nascent transcripts synthesized from the genomic dsRNA segments by the RV polymerase VP1 within the particle core. Here, from cryo-electron microscopy, x-ray crystallography, and biochemical analyses, we show that VP3 forms a stable tetrameric assembly with each subunit having a modular domain organization, which uniquely integrates five distinct enzymatic steps required for capping the transcripts. In addition to the previously known guanylyl- and methyltransferase activities, we show that VP3 exhibits hitherto unsuspected RNA triphosphatase activity necessary for initiating transcript capping and RNA helicase activity likely required for separating the RNA duplex formed transiently during endogenous transcription. From our studies, we propose a new mechanism for how VP3 inside the virion core caps the nascent transcripts exiting from the polymerase.
- Published
- 2020
49. Novel Antiretroviral Agents
- Author
-
Cambou, Mary C and Landovitz, Raphael J
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Clinical Sciences ,Infectious Diseases ,Sexually Transmitted Infections ,Clinical Trials and Supportive Activities ,Patient Safety ,Clinical Research ,Antimicrobial Resistance ,HIV/AIDS ,6.1 Pharmaceuticals ,5.1 Pharmaceuticals ,Infection ,Good Health and Well Being ,Anti-HIV Agents ,Capsid ,Capsid Proteins ,Deoxyadenosines ,Drug Discovery ,Drug Therapy ,Combination ,HIV Fusion Inhibitors ,HIV Infections ,Humans ,Organophosphates ,Piperazines ,Reverse Transcriptase Inhibitors ,HIV ,AIDS ,New antiretrovirals ,Capsid inhibitors ,Maturation inhibitors ,Nucleoside reverse transcriptase translocation inhibitors ,Anti-CD4 antibodies ,Immunology ,Virology ,Clinical sciences - Abstract
Purpose of reviewCombination antiretroviral therapy (cART) has had dramatic effects on morbidity and mortality for persons living with HIV (PLWH). Despite significant progress in treatment efficacy, tolerability, and reducing pill burden, new agents are needed to address issues of resistance, drug-drug interactions, end organ disease, and adherence. This review covers novel ART agents recently approved or in development.Recent findingsCapsid inhibitors (CAI) demonstrate high potency and potential for extended-duration dosing in pre-clinical trials. While previous maturation inhibitors (MI) were hampered by issues of drug resistance, a recent phase IIa trial for a second-generation MI demonstrated promising antiviral activity. A phase I trial to evaluate a transdermal implant of islatravir, a nucleoside reverse transcriptase translocation inhibitor (NRTTI), maintained concentrations above the target pharmacokinetic threshold at 12 weeks. The attachment inhibitor fostemsavir is available in the USA for compassionate use in multi-drug-resistant (MDR) HIV. New antiretroviral agents show promise for both extended-duration dosing and MDR HIV.
- Published
- 2020
50. TRIM34 restricts HIV-1 and SIV capsids in a TRIM5α-dependent manner.
- Author
-
Ohainle, Molly, Kim, Kyusik, Komurlu Keceli, Sevnur, Felton, Abby, Campbell, Ed, Luban, Jeremy, and Emerman, Michael
- Subjects
Antiviral Restriction Factors ,Capsid ,Capsid Proteins ,Carrier Proteins ,Clustered Regularly Interspaced Short Palindromic Repeats ,HEK293 Cells ,HIV Infections ,HIV Seropositivity ,HIV-1 ,HeLa Cells ,Humans ,Reverse Transcription ,Tripartite Motif Proteins ,Ubiquitin-Protein Ligases ,Virus Integration - Abstract
The HIV-1 capsid protein makes up the core of the virion and plays a critical role in early steps of HIV replication. Due to its exposure in the cytoplasm after entry, HIV capsid is a target for host cell factors that act directly to block infection such as TRIM5α and MxB. Several host proteins also play a role in facilitating infection, including in the protection of HIV-1 capsid from recognition by host cell restriction factors. Through an unbiased screening approach, called HIV-CRISPR, we show that the CPSF6-binding deficient, N74D HIV-1 capsid mutant is sensitive to restriction mediated by human TRIM34, a close paralog of the well-characterized HIV restriction factor TRIM5α. This restriction occurs at the step of reverse transcription, is independent of interferon stimulation, and limits HIV-1 infection in key target cells of HIV infection including CD4+ T cells and monocyte-derived dendritic cells. TRIM34 can also restrict some SIV capsids. TRIM34 restriction requires TRIM5α as knockout or knockdown of TRIM5α results in a loss of antiviral activity. Through immunofluorescence studies, we show that TRIM34 and TRIM5α colocalize to cytoplasmic bodies and are more frequently observed to be associated with infecting N74D capsids than with WT HIV-1 capsids. Our results identify TRIM34 as an HIV-1 CA-targeting restriction factor and highlight the potential role for heteromultimeric TRIM interactions in contributing to restriction of HIV-1 infection in human cells.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.