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
remove_circle_outline

Search Results (240)

Search Parameters:
Keywords = RNA virus discovery

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
48 pages, 5070 KB  
Article
Dual Inhibitory Potential of Conessine Against HIV and SARS-CoV-2: Structure-Guided Molecular Docking Analysis of Critical Viral Targets
by Ali Hazim Abdulkareem, Meena Thaar Alani, Sameer Ahmed Awad, Safaa Abed Latef Al-Meani, Mohammed Mukhles Ahmed, Elham Hazeim Abdulkareem and Zaid Mustafa Khaleel
Viruses 2025, 17(11), 1435; https://doi.org/10.3390/v17111435 - 29 Oct 2025
Viewed by 467
Abstract
Human immunodeficiency virus (HIV-1) and SARS-CoV-2 continue to co-burden global health, motivating discovery of broad-spectrum small molecules. Conessine, a steroidal alkaloid, has reported membrane-active and antimicrobial properties but remains underexplored as a dual antiviral chemotype. To interrogate conessine’s multi-target antiviral potential against key [...] Read more.
Human immunodeficiency virus (HIV-1) and SARS-CoV-2 continue to co-burden global health, motivating discovery of broad-spectrum small molecules. Conessine, a steroidal alkaloid, has reported membrane-active and antimicrobial properties but remains underexplored as a dual antiviral chemotype. To interrogate conessine’s multi-target antiviral potential against key enzymatic and entry determinants of HIV-1 and SARS-CoV-2 and to benchmark performance versus approved comparators. Eight targets were modeled: HIV-1 reverse transcriptase (RT, 3V81), protease (PR, 1HVR), integrase (IN, 3LPT), gp120–gp41 trimer (4NCO); and SARS-CoV-2 main protease (Mpro, 6LU7), papain-like protease (PLpro, 6W9C), RNA-dependent RNA polymerase (RdRp, 7BV2), spike RBD (6M0J). Ligands (conessine; positive controls: dolutegravir for HIV-1, nirmatrelvir for SARS-CoV-2) were prepared with standard protonation, minimized, and docked using AutoDock Vina v 1.2.0exhaustiveness 4; 20 poses). Binding modes were profiled in 2D/3D. Protocol robustness was verified by re-docking co-crystallized ligands (RMSD ≤ 2.0 Å). Atomistic MD (explicit TIP3P, OPLS4, 300 K/1 atm, NPT; 50–100 ns) assessed pose stability (RMSD/RMSF), pocket compaction (Rg, volume), and interaction persistence; MM/GBSA provided qualitative energy decomposition. ADMET was predicted in silico. Conessine showed coherent, hydrophobically anchored binding across both viral panels. Best docking scores (kcal·mol−1) were: HIV-1—PR −6.910, RT −6.672, IN −5.733; SARS-CoV-2—spike RBD −7.025, Mpro −5.745, RdRp −5.737, PLpro −5.024. Interaction maps were dominated by alkyl/π-alkyl packing to catalytic corridors (e.g., PR Ile50/Val82, RT Tyr181/Val106; Mpro His41/Met49; RBD L455/F486/Y489) with occasional carbon-/water-mediated H-bonds guiding orientation. MD sustained low ligand RMSD (typically ≤1.6–2.2 Å) and damped RMSF at catalytic loops, indicating pocket rigidification; MM/GBSA trends (≈ −30 to −40 kcal·mol−1, dispersion-driven) supported persistent nonpolar stabilization. Benchmarks behaved as expected: dolutegravir bound strongly to IN (−6.070) and PR (−7.319) with stable MD; nirmatrelvir was specific for Mpro and displayed weaker, discontinuous engagement at PLpro/RdRp/RBD under identical settings. ADMET suggested conessine has excellent permeability/BBB access (high logP), but liabilities include poor aqueous solubility, predicted hERG risk, and CYP2D6 substrate dependence.Conessine operates as a hydrophobic, multi-target wedge with the most favorable computed engagement at HIV-1 PR/RT and the SARS-CoV-2 spike RBD, while maintaining stable poses at Mpro and RdRp. The scaffold merits medicinal-chemistry optimization to improve solubility and de-risk cardiotoxicity/CYP interactions, followed by biochemical and cell-based validation against prioritized targets. Full article
Show Figures

Figure 1

19 pages, 1722 KB  
Review
Natural Compounds with Antiviral Activity Against Clinically Relevant RNA Viruses: Advances of the Last Decade
by David Mauricio Cañedo-Figueroa, Daniela Nahomi Calderón-Sandate, Jonathan Hernández-Castillo, Manuel Josafat Huerta-Garza, Ximena Hernández-Rodríguez, Manuel Adrián Velázquez-Cervantes, Giovanna Berenice Barrera-Aveleida, Juan Valentin Trujillo-Paez, Flor Itzel Lira-Hernández, Blanca Azucena Marquez-Reyna, Moisés León-Juárez, Ana Cristina García-Herrera, Juan Fidel Osuna-Ramos and Luis Adrián De Jesús-González
Biomolecules 2025, 15(10), 1467; https://doi.org/10.3390/biom15101467 - 16 Oct 2025
Viewed by 935
Abstract
RNA viruses remain a significant public health concern due to their rapid evolution, genetic variability, and capacity to trigger recurrent epidemics and pandemics. Over the last decade, natural products have gained attention as a valuable source of antiviral candidates, offering structural diversity, accessibility, [...] Read more.
RNA viruses remain a significant public health concern due to their rapid evolution, genetic variability, and capacity to trigger recurrent epidemics and pandemics. Over the last decade, natural products have gained attention as a valuable source of antiviral candidates, offering structural diversity, accessibility, and favorable safety profiles. This review highlights key replication mechanisms of RNA viruses and their associated therapeutic targets, including RNA-dependent RNA polymerase, viral proteases, and structural proteins mediating entry and maturation. We summarize recent advances in the identification of bioactive compounds such as flavonoids, alkaloids, terpenes, lectins, and polysaccharides that exhibit inhibitory activity against clinically relevant pathogens, including the Influenza A virus (IAV), human immunodeficiency viruses (HIV), dengue virus (DENV), Zika virus (ZIKV), and Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Special emphasis is placed on the integration of in silico screening, in vitro validation, and nanotechnology-based delivery systems that address challenges of stability, bioavailability, and specificity. Furthermore, the growing role of artificial intelligence, drug repurposing strategies, and curated antiviral databases is discussed as a means to accelerate therapeutic discovery. Despite persistent limitations in clinical translation and standardization, natural products represent a promising and sustainable platform for the development of next-generation antivirals against RNA viruses. Full article
(This article belongs to the Special Issue Molecular Mechanism and Detection of SARS-CoV-2)
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
Viewed by 1627
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

17 pages, 1832 KB  
Article
Construction and Characterization of a Vesicular Stomatitis Virus Chimera Expressing Schmallenberg Virus Glycoproteins
by Huijuan Guo, Zhigang Jiang, Jing Wang, Fang Wang, Qi Jia, Zhigao Bu, Xin Yin and Zhiyuan Wen
Vet. Sci. 2025, 12(9), 809; https://doi.org/10.3390/vetsci12090809 - 25 Aug 2025
Viewed by 887
Abstract
Schmallenberg virus (SBV) is a negative-sense RNA virus transmitted by insect vectors, causing arthrogryposis-hydranencephaly syndrome in newborn ruminants. Since its discovery in Germany and the Netherlands in 2011, SBV has rapidly spread across multiple European countries, resulting in significant economic losses in the [...] Read more.
Schmallenberg virus (SBV) is a negative-sense RNA virus transmitted by insect vectors, causing arthrogryposis-hydranencephaly syndrome in newborn ruminants. Since its discovery in Germany and the Netherlands in 2011, SBV has rapidly spread across multiple European countries, resulting in significant economic losses in the livestock industry. With the increasing global animal trade and the expanded range of insect transmission, the risk of SBV introduction into non-endemic regions is also rising. As the gold standard for serological testing, the virus neutralization test (VNT) is crucial for tracking the spread of SBV and evaluating the efficacy of vaccines. However, in non-endemic regions, the lack of local viral strains and the biosafety risks associated with introducing foreign strains pose challenges to the implementation of VNT. In this study, we employed reverse genetics techniques using vesicular stomatitis virus (VSV) to substitute the VSV G protein with the envelope glycoproteins of SBV, thereby successfully generating and rescuing the recombinant virus rVSVΔG-eGFP-SBVGPC. The recombinant virus was then thoroughly characterized in terms of SBV Gc protein expression, viral morphology, and growth kinetics. Importantly, rVSVΔG-eGFP-SBVGPC exhibited SBV-specific cell tropism and was capable of reacting with SBV-positive serum, enabling the measurement of neutralizing antibody titers. The results suggest that this recombinant virus can serve as a feasible alternative for SBV neutralization tests, with promising potential for application in serological screening and vaccine evaluation. Full article
(This article belongs to the Section Veterinary Microbiology, Parasitology and Immunology)
Show Figures

Figure 1

13 pages, 3035 KB  
Article
Topography and Nanomechanics of the Tomato Brown Rugose Fruit Virus Suggest a Fragmentation-Driven Infection Mechanism
by Péter Puskás, Katalin Salánki, Levente Herényi, Tamás Hegedűs and Miklós Kellermayer
Viruses 2025, 17(9), 1160; https://doi.org/10.3390/v17091160 - 25 Aug 2025
Viewed by 1052
Abstract
Tomato brown rugose fruit virus (ToBRFV) has been causing severe agricultural damage worldwide since its recent discovery. While related to tobacco mosaic virus, its properties and infection mechanisms are poorly understood. To uncover their structure and nanomechanics, we carried out atomic force microscopy [...] Read more.
Tomato brown rugose fruit virus (ToBRFV) has been causing severe agricultural damage worldwide since its recent discovery. While related to tobacco mosaic virus, its properties and infection mechanisms are poorly understood. To uncover their structure and nanomechanics, we carried out atomic force microscopy (AFM) measurements on individual ToBRFV particles. The virions are rod-shaped with a height and width of 9 and 30 nm, respectively. Length is widely distributed (5–1000 nm), with a mode at 30 nm. ToBRFV rods displayed a 22.4 nm axial periodicity related to structural units. Force spectroscopy revealed a Young’s modulus of 8.7 MPa, a spring constant of 0.25 N/m, and a rupture force of 1.7 nN. In the force curves a step was seen at a height of 3.3 nm, which is related to virion wall thickness. Wall thickness was also estimated by predicting coat protein structure with AlphaFold, yielding a protein with a length of 7.3 nm. Accordingly, the structural element of ToBRFv is a right circular cylinder with an equal height and diameter of ~22 nm and a wall thickness between 3.3 and 7.3 nm. Thus, at least four to nine serially linked units are required to encapsidate a single, helically organized RNA genome. Fragmentation of ToBRFV into these cylindrical structural units may result in a facilitated release of the genome and thus efficient infection. Full article
(This article belongs to the Section Viruses of Plants, Fungi and Protozoa)
Show Figures

Figure 1

16 pages, 296 KB  
Review
Human Metapneumovirus: A Narrative Review on Emerging Strategies for Prevention and Treatment
by Nicola Principi, Valentina Fainardi and Susanna Esposito
Viruses 2025, 17(8), 1140; https://doi.org/10.3390/v17081140 - 20 Aug 2025
Viewed by 1763
Abstract
Human metapneumovirus (HMPV) is a major cause of acute respiratory tract infections, particularly in infants, young children, older adults, and immunocompromised individuals. Since its discovery in 2001, the virus has been recognized for its significant clinical and socioeconomic impact. Despite extensive research, no [...] Read more.
Human metapneumovirus (HMPV) is a major cause of acute respiratory tract infections, particularly in infants, young children, older adults, and immunocompromised individuals. Since its discovery in 2001, the virus has been recognized for its significant clinical and socioeconomic impact. Despite extensive research, no licensed vaccines or antiviral therapies are currently available for HMPV. This review aims to synthesize current knowledge on HMPV prevention and treatment, and to highlight promising avenues for future interventions. Several monoclonal antibodies (mAbs) targeting conserved epitopes of the HMPV fusion (F) protein have shown strong neutralizing activity in vitro and in animal models, although none have reached clinical trials. Vaccine development, including subunit, live attenuated, vector-based, and mRNA platforms, is progressing, with some candidates showing promise in adult populations. However, data in children, especially seronegative infants, remain limited. Antiviral research has explored repurposed drugs such as ribavirin and probenecid, along with novel agents like fusion inhibitors and T-cell-based immunotherapies, though none are yet approved. The development of safe, effective interventions—especially multivalent approaches targeting multiple respiratory viruses—remains a high priority. Continued research is essential to bridge the gap between preclinical promise and clinical application and to reduce the burden of HMPV infection worldwide. Full article
(This article belongs to the Section General Virology)
15 pages, 4767 KB  
Article
First Report of the Yezo Virus Isolates Detection in Russia
by Mikhail Kartashov, Kirill Svirin, Alina Zheleznova, Alexey Yanshin, Nikita Radchenko, Valentina Kurushina, Tatyana Tregubchak, Lada Maksimenko, Mariya Sivay, Vladimir Ternovoi, Alexander Agafonov and Anastasia Gladysheva
Viruses 2025, 17(8), 1125; https://doi.org/10.3390/v17081125 - 15 Aug 2025
Cited by 1 | Viewed by 1260
Abstract
The recent discovery of the Yezo virus (YEZV) in Japan and China has raised particular concern due to its potential to cause human diseases ranging from mild febrile illnesses to severe neurological disorders. We report, for the first time, the detection of five [...] Read more.
The recent discovery of the Yezo virus (YEZV) in Japan and China has raised particular concern due to its potential to cause human diseases ranging from mild febrile illnesses to severe neurological disorders. We report, for the first time, the detection of five YEZV isolates in I. persulcatus ticks from three regions of Russia. The analysis was performed using 5318 ticks of two Ixodes genus collected in 2024 from 23 regions of Russia. The minimum infection rate of YEZV in Russia among I. persulcatus ticks was 0.12% (95% CI: 0.05–0.28). The westernmost and northernmost YEZV detection points have been recorded. YEZV isolates circulating in Russia are genetically diverse. Protein domains of Russian YEZV isolates’ genomes were characterized using HMMER, AlphaFold 3, and InterProScan. The YEZV nucleoprotein (N) of Russian isolates has a racket-shaped structure with “head” and “stalk” domains similar to those of Orthonairovirus haemorrhagiae. The Lys261–Arg261 substitution in the YEZV N Chita 2024-1 isolate occurs in the α11 structure in the region of interaction with viral RNA. Our results show that the distribution area of YEZV is much wider than previously known, provide new data on complete YEZV genomes, extend our structural insight into YEZV N, and suggest a potential target for antiviral drug development to treat YEZV infection. Full article
(This article belongs to the Special Issue Tick-Borne Viruses: Transmission and Surveillance, 2nd Edition)
Show Figures

Figure 1

27 pages, 3634 KB  
Article
Characterising the Associated Virome and Microbiota of Asian Citrus Psyllid (Diaphorina citri) in Samoa
by Kayvan Etebari, Angelika M. Tugaga, Gayatri Divekar, Olo Aleni Uelese, Sharydia S. A. Tusa, Ellis Vaega, Helmy Sasulu, Loia Uini, Yuanhang Ren and Michael J. Furlong
Pathogens 2025, 14(8), 801; https://doi.org/10.3390/pathogens14080801 - 10 Aug 2025
Viewed by 887
Abstract
The Asian citrus psyllid (Diaphorina citri) is an economically important pest of citrus as it is a vector of the bacterium (Candidatus Liberibacter asiaticus, CLas) that causes huanglongbing disease (HLB). Understanding the virome of D. citri is important for [...] Read more.
The Asian citrus psyllid (Diaphorina citri) is an economically important pest of citrus as it is a vector of the bacterium (Candidatus Liberibacter asiaticus, CLas) that causes huanglongbing disease (HLB). Understanding the virome of D. citri is important for uncovering factors that influence vector competence, to support biosecurity surveillance, and to identify candidate agents for biological control. Previous studies have identified several D. citri-associated viruses from various geographical populations of this pest. To further investigate virus diversity in this pest, high-throughput sequencing was used to analyse D. citri populations from the Samoan islands of Upolu and Savai’i. Eleven novel viruses from the Yadokariviridae, Botourmiaviridae, Nodaviridae, Mymonaviridae, Partitiviridae, Totiviridae, and Polymycoviridae were identified as well as some that corresponded to unclassified groups. In addition, microbiome analysis revealed the presence of several endosymbiotic microorganisms, including Wolbachia, as well as some plant pathogenic fungi, including Botrytis cinerea. However, the causative agent of HLB disease (CLas) was not detected in the RNA-Seq data. These findings highlight the complex and diverse microbiota associated with D. citri and suggest potential interactions and dynamics between microorganisms and psyllid-associated viruses. Further research is needed to understand the ecological significance of these discoveries, and whether the novel viruses play a role in regulating field populations of the psyllid. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Graphical abstract

23 pages, 3795 KB  
Article
Exploring Gene Expression Changes in Murine Female Genital Tract Tissues Following Single and Co-Infection with Nippostrongylus brasiliensis and Herpes Simplex Virus Type 2
by Roxanne Pillay, Pragalathan Naidoo and Zilungile L. Mkhize-Kwitshana
Pathogens 2025, 14(8), 795; https://doi.org/10.3390/pathogens14080795 - 8 Aug 2025
Viewed by 925
Abstract
Background and Aim: The immunological interactions between soil-transmitted helminths (STHs) and herpes simplex virus type 2 (HSV-2), particularly in the context of co-infection, are poorly understood. Next-generation sequencing (NGS) offers a powerful approach to explore these complex immune responses and uncover potential therapeutic [...] Read more.
Background and Aim: The immunological interactions between soil-transmitted helminths (STHs) and herpes simplex virus type 2 (HSV-2), particularly in the context of co-infection, are poorly understood. Next-generation sequencing (NGS) offers a powerful approach to explore these complex immune responses and uncover potential therapeutic targets. This study leveraged NGS and bioinformatic tools to investigate transcriptional changes and immunological pathways in female genital tract (FGT) tissues of BALB/c mice acutely infected with Nippostrongylus brasiliensis (Nb), HSV-2, or co-infected. Methods: Total RNA was harvested from FGT tissues of BALB/c mice infected with Nb, HSV-2, co-infected with both pathogens, and uninfected controls. Differentially expressed genes (DEGs) were identified by comparing uninfected versus infected FGT tissues in R using edgeR and limma packages. Immune-related genes were identified by intersecting DEGs in each group-wise comparison with immune function gene sets derived from the Mouse Genome Informatics (MGI) database. Functional and pathway enrichment analyses were performed with g: Profiler and protein–protein interaction networks were built using the STRING database and visualized with Cytoscape. Key hub genes and significant gene modules were identified using the Cytoscape plugins CytoHubba and MCODE, followed by further functional analysis of these modules. Results: NGS analysis revealed distinct gene expression profiles in response to single infection with Nb or HSV-2, with both showing significant differences when uninfected controls were compared to infected FGT tissues at a 5% false discovery rate. Notably, there were no significant differences in gene expression profiles between uninfected and co-infected FGT tissues. In the comparison of uninfected versus Nb-infected FGT tissues, 368 DEGs were identified, with 356 genes upregulated and 12 downregulated. Several immune-related genes, such as Ptprc, Ccl11, Ccr2, and Cx3cr1, were significantly altered. Pathway analysis of DEGs, hub genes, and significant modules indicated modulation of immune and defense responses. Notably, Nb infection induced a robust Th2-dominant immune response in the FGT, with downregulation of pro-inflammatory genes. This likely reflects helminth-driven modulation that may impair protective Th1 responses and highlights the systemic impact of Nb on the FGT immunity. In the comparison of uninfected versus HSV-2-infected FGT tissues, 140 DEGs were identified, with 121 upregulated and 19 downregulated. Immune-related genes, including Ldlr, Camk1d, Lrp8 and Epg5, were notably altered. HSV-2 infection led to early and predominant downregulation of immune genes, consistent with viral immune evasion strategies. In addition, functional analysis revealed enrichment in cell cycle and sterol biosynthesis pathways, suggesting that HSV-2 modulates host metabolism to support viral replication while influencing immune responses. In co-infection, no significant transcriptional changes were observed, potentially reflecting immune antagonism where Nb-induced Th2 responses may suppress HSV-2-driven Th1 immune responses. Conclusions: This preliminary study offers insights into the gene expression responses in the FGT to acute single and co-infection with Nb and HSV-2. Together, these findings reveal distinct transcriptomic changes in the FGT following Nb and HSV-2 infection, with co-infection potentially leading to immune antagonism and transcriptional equilibrium. This highlights the complex interplay between helminth- and virus-induced immune modulation in shaping FGT immunity. By leveraging NGS, this study highlights important immune-related pathways and serves as a foundation for further investigations into the mechanistic roles of DEGs in immunity to these pathogens, with potential implications for developing novel therapeutic strategies. Full article
(This article belongs to the Special Issue Immunity and Immunoregulation in Helminth Infections)
Show Figures

Graphical abstract

16 pages, 604 KB  
Review
An Update on RNA Virus Discovery: Current Challenges and Future Perspectives
by Humberto Debat and Nicolas Bejerman
Viruses 2025, 17(7), 983; https://doi.org/10.3390/v17070983 - 15 Jul 2025
Viewed by 1568
Abstract
The relentless emergence of RNA viruses poses a perpetual threat to global public health, necessitating continuous efforts in surveillance, discovery, and understanding of these pathogens. This review provides a comprehensive update on recent advancements in RNA virus discovery, highlighting breakthroughs in technology and [...] Read more.
The relentless emergence of RNA viruses poses a perpetual threat to global public health, necessitating continuous efforts in surveillance, discovery, and understanding of these pathogens. This review provides a comprehensive update on recent advancements in RNA virus discovery, highlighting breakthroughs in technology and methodologies that have significantly enhanced our ability to identify novel viruses across diverse host organisms. We explore the expanding landscape of viral diversity, emphasizing the discovery of previously unknown viral families and the role of zoonotic transmissions in shaping the viral ecosystem. Additionally, we discuss the potential implications of RNA virus discovery on disease emergence and pandemic preparedness. Despite remarkable progress, current challenges in sample collection, data interpretation, and the characterization of newly identified viruses persist. Our ability to anticipate and respond to emerging respiratory threats relies on virus discovery as a cornerstone for understanding RNA virus evolution. We address these challenges and propose future directions for research, emphasizing the integration of multi-omic approaches, advanced computational tools, and international collaboration to overcome barriers in the field. This comprehensive overview aims to guide researchers, policymakers, and public health professionals in navigating the intricate landscape of RNA virus discovery, fostering a proactive and collaborative approach to anticipate and mitigate emerging viral threats. Full article
Show Figures

Figure 1

25 pages, 3522 KB  
Article
Repurposing of Some Nucleoside Analogs Targeting Some Key Proteins of the Avian H5N1 Clade 2.3.4.4b to Combat the Circulating HPAI in Birds: An In Silico Approach
by Mohd Yasir Khan, Abid Ullah Shah, Nithyadevi Duraisamy, Mohammed Cherkaoui and Maged Gomaa Hemida
Viruses 2025, 17(7), 972; https://doi.org/10.3390/v17070972 - 10 Jul 2025
Cited by 1 | Viewed by 1058
Abstract
(1) Background: The highly pathogenic avian influenza virus H5N1 clade 2.3.4.4b is an emerging threat that poses a great risk to the poultry industry. A few human cases have been linked to the infection with this clade in many parts of the world, [...] Read more.
(1) Background: The highly pathogenic avian influenza virus H5N1 clade 2.3.4.4b is an emerging threat that poses a great risk to the poultry industry. A few human cases have been linked to the infection with this clade in many parts of the world, including the USA. Unfortunately, there are no specific vaccines or antiviral drugs that could help prevent and treat the infection caused by this virus in birds. Our major objective is to identify/repurpose some (novel/known) antiviral compounds that may inhibit viral replication by targeting some key viral proteins. (2) Methods: We used state-of-the-art machine learning tools such as molecular docking and MD-simulation methods from Biovia Discovery Studio (v24.1.0.321712). The key target proteins such as hemagglutinin (HA), neuraminidase (NA), Matrix-2 protein (M2), and the cap-binding domain of PB2 (PB2/CBD) homology models were validated through structural assessment via DOPE scores, Ramachandran plots, and Verify-3D metrics, ensuring reliable structural representations, confirming their reliability for subsequent in silico approaches. These approaches include molecular docking followed by molecular dynamics simulation for 50 nanoseconds (ns), highlighting the structural stability and compactness of the docked complexes. (3) Results: Molecular docking revealed strong binding affinities for both sofosbuvir and GS441524, particularly with the NA and PB2/CBD protein targets. Among them, GS441524 exhibited superior interaction scores and a greater number of hydrogen bonds with key functional residues of NA and PB2/CBD. The MM-GBSA binding free energy calculations further supported these findings, as GS441524 displayed more favorable binding energies compared to several known standard inhibitors, including F0045S for HA, Zanamivir for NA, Rimantadine and Amantadine for M2, and PB2-39 for PB2/CBD. Additionally, 50 ns molecular dynamics simulations highlighted the structural stability and compactness of the GS441524-PB2/CBD complex, further supporting its potential as a promising antiviral candidate. Furthermore, hydrogen bond monitor analysis over the 50 ns simulation confirmed persistent and specific interactions between the ligand and proteins, suggesting that GS441524 may effectively inhibit the NA, and PB2/CBD might potentially disrupt PB2-mediated RNA synthesis. (4) Conclusions: Our findings are consistent with previous evidence supporting the antiviral activity of certain nucleoside analog inhibitors, including GS441524, against various coronaviruses. These results further support the potential repurposing of GS441524 as a promising therapeutic candidate against H5N1 avian influenza clade 2.3.4.4b. However, further functional studies are required to validate these in silico predictions and support the inhibitory action of GS441524 against the targeted proteins of H5N1, specifically clade 2.3.4.4b. Full article
(This article belongs to the Special Issue Interplay Between Influenza Virus and Host Factors)
Show Figures

Figure 1

31 pages, 1849 KB  
Review
The Application of Single-Cell Technologies for Vaccine Development Against Viral Infections
by Hong Nhi Nguyen, Isabel O. Vanderzee and Fei Wen
Vaccines 2025, 13(7), 687; https://doi.org/10.3390/vaccines13070687 - 26 Jun 2025
Cited by 1 | Viewed by 1916
Abstract
The development of vaccines against viral infections has advanced rapidly over the past century, propelled by innovations in laboratory and molecular technologies. These advances have expanded the range of vaccine platforms beyond live-attenuated and inactivated vaccines to include recombinant platforms, such as subunit [...] Read more.
The development of vaccines against viral infections has advanced rapidly over the past century, propelled by innovations in laboratory and molecular technologies. These advances have expanded the range of vaccine platforms beyond live-attenuated and inactivated vaccines to include recombinant platforms, such as subunit proteins and virus-like particles (VLPs), and more recently, mRNA-based vaccines, while also enhancing methods for evaluating vaccine performance. Despite these innovations, a persistent challenge remains: the inherent complexity and heterogeneity of immune responses continue to impede efforts to achieve consistently effective and durable protection across diverse populations. Single-cell technologies have emerged as transformative tools for dissecting this immune heterogeneity, providing comprehensive and granular insights into cellular phenotypes, functional states, and dynamic host–pathogen interactions. In this review, we examine how single-cell epigenomic, transcriptomic, proteomic, and multi-omics approaches are being integrated across all stages of vaccine development—from infection-informed discovery to guide vaccine design, to high-resolution evaluation of efficacy, and refinement of cell lines for manufacturing. Through representative studies, we highlight how insights from these technologies contribute to the rational design of more effective vaccines and support the development of personalized vaccination strategies. Full article
(This article belongs to the Special Issue Virus-Like Particle Vaccine Development)
Show Figures

Figure 1

6 pages, 670 KB  
Opinion
Rogueing or Rescuing? A Potential New Management Approach for Roses Infected with Rose Rosette Virus
by Caleb Paslay and Akhtar Ali
Viruses 2025, 17(6), 829; https://doi.org/10.3390/v17060829 - 8 Jun 2025
Viewed by 1276
Abstract
Roses (Rosa spp.) are among the most economically and culturally significant flowering plants worldwide. However, rose cultivation faces a critical threat from rose rosette disease (RRD), which is caused by Emaravirus rosae (rose rosette virus, RRV), a negative-sense RNA virus transmitted by [...] Read more.
Roses (Rosa spp.) are among the most economically and culturally significant flowering plants worldwide. However, rose cultivation faces a critical threat from rose rosette disease (RRD), which is caused by Emaravirus rosae (rose rosette virus, RRV), a negative-sense RNA virus transmitted by the eriophyid mite Phyllocoptes fructiphilus. Current RRD management strategies mainly depend on the complete removal (rogueing) of symptomatic plants, which are effective but adds high economic and aesthetic costs. During our field and laboratory observations from 2023 to 2024, we documented that RRV often remains localized to a single cane for extended periods of time (up to 80 days) in one variety before systemic spread to other canes of the same plant. This discovery supports a proposed “rescue hypothesis”, suggesting that early pruning of symptomatic canes may prevent full-plant infection and serve as a viable alternative to rogueing under specific conditions. While preliminary, our findings offer a potentially cost-effective, less destructive management strategy. However, further research is needed to validate this hypothesis and inform integrated disease management practices are established for effective control of RRD. Full article
(This article belongs to the Section Viruses of Plants, Fungi and Protozoa)
Show Figures

Figure 1

24 pages, 1795 KB  
Review
SARS-CoV-2 Replication Revisited: Molecular Insights and Current and Emerging Antiviral Strategies
by Bryan John J. Subong and Imelda L. Forteza
COVID 2025, 5(6), 85; https://doi.org/10.3390/covid5060085 - 30 May 2025
Cited by 2 | Viewed by 3546
Abstract
The replication machinery of SARS-CoV-2 is a primary target for therapeutic intervention, and has led to significant progress in antiviral medication discovery. This review consolidates contemporary molecular insights into viral replication and rigorously assesses treatment methods at different phases of viruses’ clinical development. [...] Read more.
The replication machinery of SARS-CoV-2 is a primary target for therapeutic intervention, and has led to significant progress in antiviral medication discovery. This review consolidates contemporary molecular insights into viral replication and rigorously assesses treatment methods at different phases of viruses’ clinical development. Direct-acting antivirals, such as nucleoside analogs (e.g., remdesivir, molnupiravir) and protease inhibitors (e.g., nirmatrelvir), have shown clinical effectiveness in diminishing morbidity and hospitalization rates. Simultaneously, host-targeted medicines like baricitinib, camostat, and brequinar leverage critical host–virus interactions, providing additional pathways to reduce viral replication while possibly minimizing the development of resistance. Notwithstanding these advancements, constraints in distribution methods, antiviral longevity, and the risk of mutational evasion demand novel strategies. Promising investigational approaches encompass CRISPR-mediated RNA degradation systems, inhalable siRNA-nanoparticle conjugates, and molecular glue degraders that target host and viral proteins. Furthermore, next-generation treatments aimed at underutilized enzyme domains (e.g., NiRAN, ExoN) and host chaperone systems (e.g., TRiC complex) signify a transformative approach in antiviral targeting. The integration of high-throughput phenotypic screening, AI-driven medication repurposing, and systems virology is transforming the antiviral discovery field. An ongoing interdisciplinary endeavor is necessary to convert these findings into versatile, resistance-resistant antiviral strategies that are applicable beyond the present pandemic and in future coronavirus epidemics. Full article
Show Figures

Graphical abstract

20 pages, 2772 KB  
Article
Sex Differences in Brain Transcriptomes of Juvenile Cynomolgus Macaques
by Nadia Kabbej, Frederick J. Ashby, Alberto Riva, Paul D. Gamlin, Ronald J. Mandel, Aishwarya Kunta, Courtney J. Rouse and Coy D. Heldermon
Biomolecules 2025, 15(5), 671; https://doi.org/10.3390/biom15050671 - 6 May 2025
Viewed by 753
Abstract
Background: Behavioral, social, and physical characteristics are posited to distinguish the sexes, yet research on transcription-level sexual differences in the brain is limited. Here, we investigated sexually divergent brain transcriptomics in pre-pubertal cynomolgus macaques, a commonly used surrogate species to humans. Methods: A [...] Read more.
Background: Behavioral, social, and physical characteristics are posited to distinguish the sexes, yet research on transcription-level sexual differences in the brain is limited. Here, we investigated sexually divergent brain transcriptomics in pre-pubertal cynomolgus macaques, a commonly used surrogate species to humans. Methods: A transcriptomic profile using RNA sequencing was generated for the temporal lobe, ventral midbrain, and cerebellum of three female and three male cynomolgus macaques previously treated with an adeno-associated virus vector mix. Statistical analyses to determine differentially expressed protein-coding genes in all three lobes were conducted using DeSeq2 with a false-discovery-rate-corrected p-value of 0.05. Results: We identified target genes in the temporal lobe, ventral midbrain, and cerebellum with functions in translation, immunity, behavior, and neurological disorders that exhibited statistically significant sexually divergent expression. Conclusions: We provide potential mechanistic insights into the epidemiological differences observed between the sexes with regard to mental health and infectious diseases, such as COVID-19. Our results provide pre-pubertal information on sexual differences in non-human primate brain transcriptomics and may provide insight into health disparities between the biological sexes in humans. Full article
(This article belongs to the Section Molecular Biomarkers)
Show Figures

Figure 1

Back to TopTop