Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (203)

Search Parameters:
Keywords = capsid modeling

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 2205 KB  
Article
EPIclip: A Novel Approach for the Production of Decorated Virus-Like Particles Mediated by High-Affinity Protein Binding Partners
by Aleksandra Moleda, Olivia Bagshaw, Jonas Repkewitz, Suaad Ahmed, Attila Jakab, Pamela Gomez Jordan, Sherin Sunny, Jean-Christophe Bourdon and John Foerster
Vaccines 2026, 14(2), 129; https://doi.org/10.3390/vaccines14020129 - 28 Jan 2026
Abstract
Background: Virus-like particles (VLPs) represent key tools for the development of vaccines due to their ability to induce a potent immune response to epitopes presented on their surface. However, the decoration of VLPs with a complete heterologous protein on the surface remains a [...] Read more.
Background: Virus-like particles (VLPs) represent key tools for the development of vaccines due to their ability to induce a potent immune response to epitopes presented on their surface. However, the decoration of VLPs with a complete heterologous protein on the surface remains a bottleneck for clinical translation due to the complexity of manufacture. We present a novel platform, EPIclip™, for the decoration of VLPs mediated by high-affinity protein binding partners, colicin E7 (ColE7) and immunity protein 7 (Im7), within a single prokaryotic host. We evaluate this approach using a modified hepatitis B core capsid protein and IL-31 as a model epitope. IL-31 is a prominent therapeutic target for the development of pruritic diseases. Methods: We explore the design and development of the platform, including the use of T-cell-stimulating peptides. We demonstrate several small-scale purification methods for the candidate VLP, as well as morphological analysis by transmission electron microscopy (TEM). Further, we vaccinate mice with IL-31-displaying VLPs to evaluate immunogenicity and the ability to prevent IL-31-induced pruritus in vivo. Results: Our results demonstrate that decorated VLPs dosed in mice elicit an IgG response against IL-31 with at least six months of durability. In addition, IL-31-displaying VLPs suppress the development of IL-31-induced pruritus, confirming in vivo target neutralisation. Notably, IL-31-displaying VLPs induce a strong T-cell response against the VLP capsid but not against the cytokine, confirming a B-cell-biased immune response and the absence of detrimental autoreactive T cells. We further demonstrate the translation of this system with an additional virus capsid: tomato aspermy virus (TAV). Conclusions: Taken together, the novel EPIclip™ platform may represent a promising therapeutic approach for pruritic diseases. Additionally, this modular system could be adapted for a wide range of research as well as human and veterinary therapeutic applications. Full article
(This article belongs to the Section Vaccine Design, Development, and Delivery)
Show Figures

Figure 1

22 pages, 4979 KB  
Article
Investigating the Potential Role of Capsaicin in Facilitating the Spread of Coxsackievirus B3 via Extracellular Vesicles
by Shruti Chatterjee, Ramina Kordbacheh, Haylee Tilley, Devin Briordy, Richard T. Waldron, William D. Cutts, Jayden Aleman, Alexis Cook, Raeesa Dhanji, Lok-Yin Roy Wong, Stephen J. Pandol, Brandon J. Kim, DeLisa Fairweather and Jon Sin
Int. J. Mol. Sci. 2026, 27(2), 661; https://doi.org/10.3390/ijms27020661 - 9 Jan 2026
Viewed by 199
Abstract
Coxsackievirus B3 (CVB3) is a picornavirus that causes systemic inflammatory diseases including myocarditis, pericarditis, pancreatitis, and meningoencephalitis. We have previously reported that CVB3 induces mitochondrial fission and mitophagy while inhibiting lysosomal degradation by blocking autophagosome-lysosome fusion. This promotes the release of virus-laden mitophagosomes [...] Read more.
Coxsackievirus B3 (CVB3) is a picornavirus that causes systemic inflammatory diseases including myocarditis, pericarditis, pancreatitis, and meningoencephalitis. We have previously reported that CVB3 induces mitochondrial fission and mitophagy while inhibiting lysosomal degradation by blocking autophagosome-lysosome fusion. This promotes the release of virus-laden mitophagosomes from host cells as infectious extracellular vesicles (EVs), enabling non-lytic viral egress. Transient receptor potential vanilloid 1 (TRPV1), a heat and capsaicin-sensitive cation channel, regulates mitochondrial dynamics by inducing mitochondrial membrane depolarization and fission. In this study, we found that TRPV1 activation by capsaicin dramatically enhances CVB3 egress from host cells via EVs. Released EVs revealed increased levels of viral capsid protein VP1, mitochondrial protein TOM70, and fission protein phospho-DRP1. Moreover, these EVs were enriched in heat shock protein HSP70, suggesting its role in facilitating infectious EV release from cells. Furthermore, TRPV1 inhibition with capsazepine and SB-366791 significantly reduced viral infection in vitro. Our in vivo studies also found that SB-366791 significantly mitigates pancreatic damage and reduces viral titers in a mouse model of CVB3 pancreatitis. Given the lack of understanding regarding factors that contribute to diverse clinical manifestations of CVB3, our study highlights capsaicin and TRPV1 as potential exacerbating factors that facilitate CVB3 dissemination via mitophagy-derived EVs. Full article
Show Figures

Figure 1

20 pages, 3781 KB  
Article
Preclinical Assessment of a New Virus-like Particle-Based Quadrivalent Human Papillomavirus Vaccine in Animal Models
by Hajar Mohammadi Barzelighi, Zahra Naderi Saffar, Erfan Pakatchian, Mohammad Taqavian, Babak Javadimehr, Mansooreh Safaeian, Payam Abbaszadeh and Hasan Jalili
Vaccines 2026, 14(1), 66; https://doi.org/10.3390/vaccines14010066 - 5 Jan 2026
Viewed by 446
Abstract
Background: A quadrivalent HPV vaccine (BPV) has been developed to prevent diseases caused by HPV types 6, 11, 16, and 18 for the first time in Iran. The BPV is composed of the papillomavirus major capsid protein L1, which serves as the primary [...] Read more.
Background: A quadrivalent HPV vaccine (BPV) has been developed to prevent diseases caused by HPV types 6, 11, 16, and 18 for the first time in Iran. The BPV is composed of the papillomavirus major capsid protein L1, which serves as the primary target in the design of the prophylactic HPV vaccines. To enhance immunogenicity, BPV was formulated with an amorphous aluminum hydroxy phosphate sulfate adjuvant. Methods: The immunogenicity and safety of BPV were assessed through analyses of both humoral and cell-mediated immunity, single and repeated doses, and reproductive effects using animal models. Results: Acute toxicity assessments showed no abnormalities in ophthalmic examinations, biochemical profiles, hematological parameters, and gross pathology findings. Additionally, no mortality or abnormal clinical signs were observed during a 90-day repeated-dose toxicity study. While some inflammatory reactions were noted at the injection sites and in the liver tissues of BPV-treated groups, these reactions were resolved by day 90 after the initial BPV administration. Furthermore, no signs of toxicity were detected in F1 offspring, and no adverse effects were identified in maternal reproductive performance, fertility, or hematological or biochemical parameters throughout the study duration. The BPV candidate successfully induced T-cell proliferation and increased the proportions of CD3+ CD4+ and CD3+ CD8+ T cells. It also stimulated the secretion of both interferon gamma (IFN-γ) and interleukin-4 (IL-4) cytokines in splenocytes isolated from animal models after the third dose. Moreover, anti-HPV L1 IgG antibody production was confirmed on day 14 after administration of each of the three BPV vaccine doses. Conclusions: The findings suggest that BPV is a vaccine candidate that stimulates both cellular and humoral immunity and demonstrate its safety profile in animal models. Full article
(This article belongs to the Section Human Papillomavirus Vaccines)
Show Figures

Graphical abstract

20 pages, 1661 KB  
Article
Structure-Guided Engineering of Protein VP2 from Epizootic Hemorrhagic Disease Virus Maximizes Production and Confers Complete Protection as Subunit Vaccine
by Samuel Jurado, Luis Jiménez-Cabello, María del Carmen Nuñez, Sergio Utrilla-Trigo, Eva Calvo-Pinilla, Iván Mazuecos-Aragonés, José Ramón Gutierrez, Ana Falcón, Javier Ortego and José M. Escribano
Vaccines 2026, 14(1), 7; https://doi.org/10.3390/vaccines14010007 - 20 Dec 2025
Viewed by 511
Abstract
Epizootic hemorrhagic disease (EHD) is an important livestock disease caused by Epizootic hemorrhagic disease virus (EHDV). The recent incursion and wide distribution of EHDV in Europe have increased the need for effective vaccine candidates. Background/Objectives: The VP2 protein of EHDV forms the outer [...] Read more.
Epizootic hemorrhagic disease (EHD) is an important livestock disease caused by Epizootic hemorrhagic disease virus (EHDV). The recent incursion and wide distribution of EHDV in Europe have increased the need for effective vaccine candidates. Background/Objectives: The VP2 protein of EHDV forms the outer capsid layer of the virion and is essential for viral assembly and host cell entry. Owing to its antigenic properties, VP2 represents a major target for vaccine development. However, the recombinant production of VP2 is limited by low stability and poor yields, representing a significant barrier for the generation of safe and effective subunit vaccines. Methods: To overcome these limitations, the VP2 protein from EHDV serotype 8 (EHDV-8) was rationally engineered with targeted modifications at both the amino and carboxyl termini of its coding sequence. Recombinant expression was performed using a baculovirus vector-mediated system in Trichoplusia ni pupae (CrisBio® technology), employed as living biofactories. Results: The engineering of VP2 resulted in up to a tenfold increase in protein yields compared with the wild-type sequence, while maintaining the trimeric structural integrity of the recombinant protein. Both wild-type and engineered VP2 protein variants were formulated and used to immunize IFNAR(−/−) mice, a model susceptible to EHDV infection. Both engineered and wild-type VP2 formulations elicited comparable neutralizing antibody responses in vaccinated animals. Furthermore, immunization with either formulation conferred full protection against lethal EHDV-8 challenge. Conclusions: In this work, we demonstrated that the rational engineering of the VP2 protein significantly improved recombinant expression yields in a baculovirus-based system without compromising structural integrity or immunogenicity. These findings additionally demonstrate the feasibility of producing high-quality VP2 antigens in T. ni pupae using CrisBio® technology and support their potential application in the development of subunit vaccines against EHDV. Full article
Show Figures

Figure 1

17 pages, 1312 KB  
Article
Avian Immunoglobulin Y Antibodies Targeting the Protruding or Shell Domain of Norovirus Capsid Protein Neutralize Norovirus Replication in the Human Intestinal Enteroid System
by Ming Xia, Mohamed Ichou, Mathew Landivar, Peng Zhou, Sai Navya Vadlamudi, Alice Leruth, Charlotte Nyblade, Paul Cox, Lijuan Yuan, Julius Goepp and Ming Tan
Vaccines 2025, 13(12), 1228; https://doi.org/10.3390/vaccines13121228 - 5 Dec 2025
Viewed by 711
Abstract
Background: Norovirus is a leading cause of epidemic acute gastroenteritis worldwide, associated with significant morbidity, mortality, and economic loss. Despite its global impact, no licensed vaccine is currently available, and vaccine development remains challenging. Methods: We explored avian immunoglobulin Y (IgY) antibodies as [...] Read more.
Background: Norovirus is a leading cause of epidemic acute gastroenteritis worldwide, associated with significant morbidity, mortality, and economic loss. Despite its global impact, no licensed vaccine is currently available, and vaccine development remains challenging. Methods: We explored avian immunoglobulin Y (IgY) antibodies as a low-cost countermeasure against norovirus infection. We generated recombinant protruding (P) domain proteins from the capsid protein (VP1) of noroviruses, representing two GII.4 variants and the GII.6 genotype. These were combined into a single immunogen to immunize laying hens to produce norovirus VP1-specific IgY antibodies. Results: Immunization of laying hens with the P domain proteins elicited high-titer (>1:450,000) P domain-specific IgY antibodies. The yolk-derived IgY effectively inhibited binding of various norovirus P particles to their histo-blood group antigen ligands, with 50% blocking titers (BT50) up to 1:8533 against homotypic GII.4 and 1:667 against heterotypic G1.1 Norwalk virus P particles. Importantly, the IgY neutralized replication of GII.4 norovirus in the human intestinal enteroid (HIE) system at a high titer of over 1:2500, equivalent to 0.70 µg/mL of total IgY. We also produced norovirus shell (S) domain proteins and corresponding IgY antibodies, which neutralized GII.4 norovirus replication in the HIE model at a titer of ~1:800, equivalent to 2.98 µg/mL of total IgY. This provides the first evidence that the S domain contains neutralizing epitopes. Conclusions: Our findings support the potential of IgY targeting norovirus P or S domains as a scalable, cost-effective strategy for preventing norovirus infection and disease. Full article
(This article belongs to the Special Issue New Approaches to Vaccine Development and Delivery)
Show Figures

Figure 1

19 pages, 2271 KB  
Article
Plasmonic Nanopore Sensing to Probe the DNA Loading Status of Adeno-Associated Viruses
by Scott Renkes, Steven J. Gray, Minjun Kim and George Alexandrakis
Chemosensors 2025, 13(12), 418; https://doi.org/10.3390/chemosensors13120418 - 4 Dec 2025
Cited by 1 | Viewed by 1016
Abstract
Adeno-associated viruses (AAVs) are a leading vector for gene therapy, yet their clinical utility is limited by the lack of robust quality control methods to distinguish between empty (AAVempty), partially loaded (AAVpartial), and fully DNA loaded (AAVfull) [...] Read more.
Adeno-associated viruses (AAVs) are a leading vector for gene therapy, yet their clinical utility is limited by the lack of robust quality control methods to distinguish between empty (AAVempty), partially loaded (AAVpartial), and fully DNA loaded (AAVfull) capsids. Current analytical techniques provide partial insights but remain limited in sensitivity, throughput, or resolution. Here we present a multimodal plasmonic nanopore sensor that integrates optical trapping with electrical resistive-pulse sensing to characterize AAV9 capsids at the single-particle level in tens of μL sample volumes and fM range concentrations. As a model system, we employed AAV9 capsids not loaded with DNA, capsids loaded with a self-complementary 4.7 kbp DNA (AAVscDNA), and ones loaded with single-stranded 4.7 kbp DNA (AAVssDNA). Ground-truth validation was performed with analytical ultracentrifugation (AUC). Nanosensor data were acquired concurrently for optical step changes (occurring at AAV trapping and un-trapping) both in transmittance and reflectance geometries, and electrical nanopore resistive pulse signatures, making for a total of five data dimensions. The acquired data was then filtered and clustered by Gaussian mixture models (GMMs), accompanied by spectral clustering stability analysis, to successfully separate between AAV species based on their DNA load status (AAVempty, AAVpartial, AAVfull) and DNA load type (AAVscDNA versus AAVssDNA). The motivation for quantifying the AAVempty and AAVpartial population fractions is that they reduce treatment efficacy and increase immunogenicity. Likewise, the motivation to identify AAVscDNA population fractions is that these have much higher transfection rates. Importantly, the results showed that the nanosensor could differentiate between AAVscDNA and AAVssDNA despite their identical masses. In contrast, AUC could not differentiate between AAVscDNA and AAVssDNA. An equimolar mixture of AAVscDNA, AAVssDNA and AAVempty was also measured with the sensor, and the results showed the expected population fractions, supporting the capacity of the method to differentiate AAV load status in heterogeneous solutions. In addition, less common optical and electrical signal signatures were identified in the acquired data, which were attributed to debris, rapid entry re-entry to the optical trap, or weak optical trap exits, representing critical artifacts to recognize for correct interpretation of the data. Together, these findings establish plasmonic nanopore sensing as a promising platform for quantifying AAV DNA loading status and genome type with the potential to extend ultra-sensitive single-particle characterization beyond the capabilities of existing methods. Full article
(This article belongs to the Special Issue Electrochemical Sensors Based on Various Materials)
Show Figures

Figure 1

9 pages, 393 KB  
Review
Reassessing Viral Origins: From Escaped Genes to Degenerated Microbes
by Peter Borger
Pathogens 2025, 14(12), 1205; https://doi.org/10.3390/pathogens14121205 - 26 Nov 2025
Viewed by 874
Abstract
Three main hypotheses have been proposed to explain the origin of viruses: the exogenisation (escape) hypothesis, suggesting that mobile genetic elements gained infectivity and autonomy; the degeneration hypothesis, proposing that viruses arose through gene loss from more complex, possibly cellular ancestors; and the [...] Read more.
Three main hypotheses have been proposed to explain the origin of viruses: the exogenisation (escape) hypothesis, suggesting that mobile genetic elements gained infectivity and autonomy; the degeneration hypothesis, proposing that viruses arose through gene loss from more complex, possibly cellular ancestors; and the virus-first hypothesis, which argues that viruses are ancient, pre-cellular entities. This review evaluates these models in light of molecular, structural, and ecological evidence. Key considerations include the lack of homologues for many viral proteins, the presence of giant DNA viruses with extensive gene repertoires, the conservation of capsid structures across diverse viruses, and the universal dependence of viruses on living hosts. Also discussed is the vast diversity of the global virosphere revealed by recent metaviromic studies, particularly in marine ecosystems, where viruses play key roles in structuring microbial communities and driving biogeochemical cycles. Such findings highlight that viruses are integral components of biological systems rather than merely parasitic outliers. Although no single hypothesis fully explains the origin of all viruses, their extraordinary genetic and functional complexity suggests a unified evolutionary theory may forever remain elusive. Rather, understanding the origins of viruses requires integrating genomic traits with ecological roles across their wide diversity. Full article
Show Figures

Figure 1

25 pages, 1627 KB  
Review
Resveratrol as an Adjunct Antiviral Agent in Pediatric Viral Infections: A Review on Mechanistic Insights and Gut Microbiota Modulation
by Roberta Leonardi, Manuela Lo Bianco, Salvatore Spinello, Pasqua Betta, Caterina Gagliano, Vittorio Calabrese, Agata Polizzi and Giulia Malaguarnera
Int. J. Mol. Sci. 2025, 26(23), 11341; https://doi.org/10.3390/ijms262311341 - 24 Nov 2025
Cited by 1 | Viewed by 969
Abstract
Pediatric viral infections impose a heavy burden on child health, often worsened by infection-induced gut dysbiosis. Resveratrol, a natural polyphenol with antiviral, anti-inflammatory, and microbiota-modulating properties, has been proposed to interrupt this pathogenic feedback. To our knowledge, this is the first narrative review [...] Read more.
Pediatric viral infections impose a heavy burden on child health, often worsened by infection-induced gut dysbiosis. Resveratrol, a natural polyphenol with antiviral, anti-inflammatory, and microbiota-modulating properties, has been proposed to interrupt this pathogenic feedback. To our knowledge, this is the first narrative review focused on resveratrol’s antiviral activity in pediatric viral infections, concurrently evaluating its impact on the gut microbiota and their interrelationship. We synthetized preclinical and the limited available pediatric clinical data regarding resveratrol’s effect on SARS-CoV-2, respiratory syncytial virus, influenza, rotavirus, and norovirus, extracting information on the models, routes of administration, dosages, mechanisms, and outcomes. Resveratrol interferes with viral lifecycles via diverse mechanisms (modulation of host signaling cascades, capsid or structural protein interactions, and suppression of pro-viral chaperones) while concurrently reshaping the gut microbiota (reducing opportunistic taxa and enriching beneficial genera such as Bifidobacterium and Lactobacillus) leading to improved short-chain fatty acid profiles, barrier integrity, and dampened inflammation. Intranasal resveratrol in children shows clinical benefit, while oral use is underexplored and limited by poor bioavailability; adult data hint at supportive microbiome and anti-inflammatory effects if the delivery is optimized. These dual antiviral and microbiome-directed effects position resveratrol as a promising adjunct in pediatric viral disease management, though well-powered pediatric clinical trials are needed to define dosages, delivery strategies, and the contribution of microbiota-mediated synergy. Full article
Show Figures

Figure 1

47 pages, 1332 KB  
Review
Base and Prime Editing for Inherited Retinal Diseases: Delivery Platforms, Safety, Efficacy, and Translational Perspectives
by Haoliang Zhang, Yuxuan Li, Jiajie Li, Xiaosa Li and Tong Li
Pharmaceutics 2025, 17(11), 1405; https://doi.org/10.3390/pharmaceutics17111405 - 30 Oct 2025
Viewed by 3131
Abstract
Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous spectrum of disorders that lead to progressive and irreversible vision loss. Gene therapy is the most promising emerging treatment for IRDs. While gene augmentation strategies have demonstrated clinical benefit and results within the [...] Read more.
Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous spectrum of disorders that lead to progressive and irreversible vision loss. Gene therapy is the most promising emerging treatment for IRDs. While gene augmentation strategies have demonstrated clinical benefit and results within the first approved ocular gene therapy, their application is restricted by adeno-associated virus (AAV) packaging capacity and limited efficacy for dominant mutations. Recent breakthroughs in precision genome editing, particularly base editing (BE) and prime editing (PE), have provided alternatives capable of directly correcting pathogenic variants. BE enables targeted single-nucleotide conversions, whereas PE further allows for precise insertions and deletions, both circumventing the double-strand DNA cleavage or repair processes typically induced by conventional CRISPR–Cas editing systems, thereby offering advantages in post-mitotic retinal cells. Preclinical investigations across murine and non-human primate models have demonstrated the feasibility, molecular accuracy, and preliminary safety profiles of these platforms in targeting IRD-associated mutations. However, critical challenges remain before clinical application can be realized, including limited editing efficiency in photoreceptors, interspecies variability in therapeutic response, potential risks of off-target effects, and barriers in large-scale vector manufacturing. Moreover, the delivery of genome editors to the outer retina remains suboptimal, prompting intensive efforts in capsid engineering and the development of non-viral delivery systems. This review synthesizes the current progress in BE and PE optimization, highlights innovations in delivery platforms that encompass viral and emerging non-viral systems and summarizes the major barriers to clinical translation. We further discuss AI-driven strategies for the rational design of BE/PE systems, thereby outlining their future potential and perspectives in the treatment of IRDs. Full article
(This article belongs to the Special Issue Ophthalmic Drug Delivery, 3rd Edition)
Show Figures

Graphical abstract

11 pages, 604 KB  
Review
HIV Therapy: The Latest Developments in Antiviral Drugs—A Scoping Review
by Francisco Fanjul, Meritxell Gavalda, Antoni Campins, Adria Ferré, Luisa Martín, María Peñaranda, Mari Ángeles Ribas, Elena Pastor-Ramon, Sophia Pinecki and Melchor Riera
Biomedicines 2025, 13(11), 2629; https://doi.org/10.3390/biomedicines13112629 - 27 Oct 2025
Viewed by 3599
Abstract
Background: Major advances in antiretroviral therapy (ART) have transformed HIV into a chronic condition, yet drug resistance, long-term toxicities, adherence challenges, and persistent viral reservoirs continue to drive innovation. Objectives: To map and synthesize recent developments in anti-HIV drugs and delivery platforms with [...] Read more.
Background: Major advances in antiretroviral therapy (ART) have transformed HIV into a chronic condition, yet drug resistance, long-term toxicities, adherence challenges, and persistent viral reservoirs continue to drive innovation. Objectives: To map and synthesize recent developments in anti-HIV drugs and delivery platforms with a focus on (i) new molecules in clinical development and (ii) novel mechanisms of action, following a scoping review framework aligned with PRISMA-ScR. Sources: We interrogated PubMed, Embase.com, Web of Science, and Scopus (January 2020–September 2025) and screened abstracts from CROI, IAS/AIDS, IDWeek, and HIV Glasgow (2023–2025). Content: The evidence base underscores capsid inhibition (lenacapavir) for multidrug-resistant HIV and its expansion into prevention, long-acting intramuscular maintenance with cabotegravir/rilpivirine, maturation inhibitors (zabofiravir), and attachment inhibition with fostemsavir. Broadly neutralizing antibodies (bNAbs) can sustain ART-free suppression in selected individuals. Ultra-long-acting delivery systems are advancing toward translational evaluation. Summary: The pipeline is diversifying toward less frequent dosing, new targets, and combination strategies. Successful and ethical implementation will require resistance-informed selection, equitable access, and reimagined healthcare delivery models that accommodate long-acting technologies. Full article
(This article belongs to the Special Issue HIV Therapy: The Latest Developments in Antiviral Drugs)
Show Figures

Figure 1

15 pages, 3697 KB  
Article
Virus-like Particles Formed by the Coat Protein of the Single-Stranded RNA Phage PQ465 as a Carrier for Antigen Presentation
by Egor A. Vasyagin, Eugenia S. Mardanova and Nikolai V. Ravin
Molecules 2025, 30(20), 4056; https://doi.org/10.3390/molecules30204056 - 11 Oct 2025
Cited by 1 | Viewed by 993
Abstract
Virus-like particles (VLPs) formed as a result of self-assembly of viral capsid proteins are widely used as a platform for antigen presentation in vaccine development. However, since the inclusion of a foreign peptide into the capsid protein can alter its spatial structure and [...] Read more.
Virus-like particles (VLPs) formed as a result of self-assembly of viral capsid proteins are widely used as a platform for antigen presentation in vaccine development. However, since the inclusion of a foreign peptide into the capsid protein can alter its spatial structure and interfere with VLP assembly, such insertions are usually limited to short peptides. In this study, we have demonstrated the potential of capsid protein (CP) of single-stranded RNA phage PQ465 to present long peptides using green fluorescent protein (GFP) as a model. GFP was genetically linked to either the N- or C-terminus of PQ465 CP. Hybrid proteins were expressed in Escherichia coli and Nicotiana benthamiana plants. Spherical virus-like particles (~35 nm according to transmission electron microscopy) were successfully formed by both N- and C-terminal fusions expressed in E. coli, and by plant-produced CP with GFP fused to the C-terminus. ELISA revealed that GFP in VLPs was accessible for specific antibodies suggesting that it is exposed on the surface of PQ465-GFP particles. VLPs carrying GFP were recognized by anti-CP antibodies with less efficiency than VLPs formed by empty CP, which indicates shielding of the CP core in PQ465-GFP particles. Therefore, PQ465 CP can be used as a chimeric VLP platform for the display of relatively large protein antigens, which can operate in bacterial and plant expression systems. Full article
(This article belongs to the Special Issue Recent Advances in Peptide Assembly and Bioactivity)
Show Figures

Figure 1

18 pages, 4107 KB  
Article
Intertypic Recombination Between Coxsackievirus A16 and Enterovirus A71 Structural and Non-Structural Genes Modulates Virulence and Protection Efficacy
by Hooi Yee Chang, Han Kang Tee, Kien Chai Ong, Kartini Jasni, Syahril Abdullah, I.-Ching Sam and Yoke Fun Chan
Vaccines 2025, 13(10), 1017; https://doi.org/10.3390/vaccines13101017 - 29 Sep 2025
Viewed by 1107
Abstract
Background/Objectives: Enterovirus A71 (EV-A71) and coxsackievirus A16 (CVA16) are major causative agents of hand, foot and mouth disease (HFMD), often co-circulating and occasionally undergoing genetic recombination. While natural recombinants often involve genomic regions encoding non-structural proteins, their effects on replication and pathogenesis [...] Read more.
Background/Objectives: Enterovirus A71 (EV-A71) and coxsackievirus A16 (CVA16) are major causative agents of hand, foot and mouth disease (HFMD), often co-circulating and occasionally undergoing genetic recombination. While natural recombinants often involve genomic regions encoding non-structural proteins, their effects on replication and pathogenesis remain unclear. Methods: To address this, four chimera viruses (Chi-CCE, Chi-ECE, Chi-EEC, and Chi-CEC) were constructed with 5′UTR, capsid P1, and non-structural P2 and P3 genes, from CVA16 (denoted as C) or EV-A71 (denoted as E). These chimeras were tested for replication kinetics and cytopathic effects in rhabdomyosarcoma cells while in vivo virulence and protection efficacy were evaluated using a newborn BALB/c mouse model. Results: All chimeric viruses remained viable and exhibited higher replication than CVA16. In vivo, all chimeric viruses were avirulent except Chi-CCE and CVA16, which showed high virulence and viral titres in the brains and limbs of infected newborn mice. This suggests that 5′UTR and capsid P1 genes of CVA16 are critical genetic determinants of virulence. Notably, only the anti-inflammatory cytokine IL-10 was elevated, suggesting potential immune modulation during infection. Inactivated Chi-CCE immunisation conferred 100% protection against lethal CVA16 or mouse-adapted EV-A71 challenge revealing its potential as a bivalent vaccine candidate. Conclusions: Our study demonstrates that recombination between CVA16 and EV-A71 influences viral virulence and protection efficacy with implications for future development of multivalent vaccines. Full article
(This article belongs to the Section Vaccines Against Tropical and Other Infectious Diseases)
Show Figures

Figure 1

15 pages, 3995 KB  
Article
Screening of Single-Domain Antibodies to Adeno-Associated Viruses with Cross-Serotype Specificity and a Wide pH Tolerance
by Hailing Guo, Shuo Wang, Lujin Feng, Weiwei Xu, Jiandong Zhang, Xiaoju Zhou and Ningning Ma
Viruses 2025, 17(10), 1289; https://doi.org/10.3390/v17101289 - 23 Sep 2025
Viewed by 3690
Abstract
Adeno-associated virus (AAV) vectors are the preferred gene delivery tool in gene therapy owing to their safety, long-term gene expression, broad tissue tropism, and low immunogenicity. Affinity ligands that can bind multiple AAV serotypes endure harsh clean-in-place (CIP) conditions and are critical for [...] Read more.
Adeno-associated virus (AAV) vectors are the preferred gene delivery tool in gene therapy owing to their safety, long-term gene expression, broad tissue tropism, and low immunogenicity. Affinity ligands that can bind multiple AAV serotypes endure harsh clean-in-place (CIP) conditions and are critical for industrial-scale purification. However, current ligands lack broad serotype recognition and adequate alkaline stability, which limits their reusability in large-scale manufacturing. In this study, we employed a competitive biopanning strategy to isolate a single-domain antibody (VHH) that simultaneously binds AAV2, AAV8, and AAV9. The VHH retained structural integrity and binding activity after exposure to 0.1 M NaOH, demonstrating robust alkaline stability. Structural modeling revealed that the VHH primarily recognizes the DE loop region of the VP3 capsid protein across the three serotypes, explaining its cross-serotype reactivity. Affinity chromatography using the VHH yielded infectious AAV particles, confirming its potential for downstream processing. This strategy provides a versatile platform for developing high-performance AAV affinity ligands and may be extended to other viral vector systems. Full article
(This article belongs to the Section General Virology)
Show Figures

Graphical abstract

17 pages, 1606 KB  
Article
Structural Insights into the Nuclear Import of Haliotid Herpesvirus 1 Large Tegument Protein Homologue
by Babu Kanti Nath, Crystall M. D. Swarbrick, Renate H. M. Schwab, Daryl Ariawan, Ole Tietz, Jade K. Forwood and Subir Sarker
Viruses 2025, 17(9), 1279; https://doi.org/10.3390/v17091279 - 20 Sep 2025
Cited by 1 | Viewed by 849
Abstract
Abalone are highly susceptible to haliotid herpesvirus 1 (HaHV1), the causative agent of abalone viral ganglioneuritis (AVG), a re-emerging disease responsible for significant mortality events in both wild and farmed populations. Currently, there are no effective antiviral treatments or preventive measures available against [...] Read more.
Abalone are highly susceptible to haliotid herpesvirus 1 (HaHV1), the causative agent of abalone viral ganglioneuritis (AVG), a re-emerging disease responsible for significant mortality events in both wild and farmed populations. Currently, there are no effective antiviral treatments or preventive measures available against HaHV1, which is partly due to the limited understanding of the immune responses and viral pathogenesis in this non-model marine invertebrate. This highlights the urgent need for novel intervention strategies, including investigations into the molecular mechanisms underlying HaHV1 infection. In other herpesviruses, the large tegument protein UL36 plays a crucial role in transporting the viral capsid to the host cell’s nuclear pore complex (NPC), mediated by N-terminal nuclear localization signals (NLSs). However, the nuclear import mechanism of UL36 homologue (UL36h) in HaHV1 remains largely uncharacterized. In this study, we identified and functionally characterized the NLS motif within HaHV1 UL36h and elucidated its interactions with the importin alpha (IMPα) nuclear import receptor. Through a combination of high-resolution crystallography and quantitative binding assays, we determined the key residues responsible for binding to IMPα and demonstrated isoform-specific variations in binding affinity. Our biochemical and structural analyses confirmed key interactions within the NLS that are essential for IMPα interactions. These findings advance our molecular understanding of HaHV1 host interactions and pave the way for the development of targeted antiviral strategies against abalone herpesvirus infection. Full article
Show Figures

Figure 1

44 pages, 2436 KB  
Review
Antiviral Strategies Targeting Enteroviruses: Current Advances and Future Directions
by Michelle Felicia Lee, Seng Kong Tham and Chit Laa Poh
Viruses 2025, 17(9), 1178; https://doi.org/10.3390/v17091178 - 28 Aug 2025
Cited by 1 | Viewed by 2586
Abstract
Enteroviruses, a diverse genus within the Picornaviridae family, are responsible for a wide range of human infections, including hand, foot, and mouth disease, respiratory disease, aseptic meningitis, encephalitis, myocarditis, and acute flaccid paralysis. Despite their substantial global health burden and the frequent emergence [...] Read more.
Enteroviruses, a diverse genus within the Picornaviridae family, are responsible for a wide range of human infections, including hand, foot, and mouth disease, respiratory disease, aseptic meningitis, encephalitis, myocarditis, and acute flaccid paralysis. Despite their substantial global health burden and the frequent emergence of outbreaks, no specific antiviral therapies are currently approved for clinical use against non-polio enteroviruses. This review provides a comprehensive overview of the current landscape of antiviral strategies targeting enteroviruses, including direct-acting antivirals such as capsid binders, protease inhibitors, and viral RNA polymerase inhibitors. We also examine the potential of host-targeting agents that interfere with virus–host interactions essential for replication. Emerging strategies such as immunotherapeutic approaches, RNA interference, CRISPR-based antivirals, and peptide-based antivirals are also explored. Furthermore, we address key challenges, including viral diversity, drug resistance, and limitations in preclinical models. By highlighting recent advances and ongoing efforts in antiviral development, this review aims to guide future research and accelerate the discovery of effective therapies against enterovirus infections. Full article
(This article belongs to the Special Issue Picornavirus Evolution, Host Adaptation and Antiviral Strategies)
Show Figures

Graphical abstract

Back to TopTop