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

Search Results (267)

Search Parameters:
Keywords = fusion glycoprotein

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 8583 KiB  
Article
Development and Immunogenic Evaluation of a Recombinant Vesicular Stomatitis Virus Expressing Nipah Virus F and G Glycoproteins
by Huijuan Guo, Renqiang Liu, Dan Pan, Yijing Dang, Shuhuai Meng, Dan Shan, Xijun Wang, Jinying Ge, Zhigao Bu and Zhiyuan Wen
Viruses 2025, 17(8), 1070; https://doi.org/10.3390/v17081070 - 31 Jul 2025
Viewed by 135
Abstract
Nipah virus (NiV) is a highly pathogenic bat-borne zoonotic pathogen that poses a significant threat to human and animal health, with fatality rates exceeding 70% in some outbreaks. Despite its significant public health impact, there are currently no licensed vaccines or specific therapeutics [...] Read more.
Nipah virus (NiV) is a highly pathogenic bat-borne zoonotic pathogen that poses a significant threat to human and animal health, with fatality rates exceeding 70% in some outbreaks. Despite its significant public health impact, there are currently no licensed vaccines or specific therapeutics available. Various virological tools—such as reverse genetics systems, replicon particles, VSV-based pseudoviruses, and recombinant Cedar virus chimeras—have been widely used to study the molecular mechanisms of NiV and to support vaccine development. Building upon these platforms, we developed a replication-competent recombinant vesicular stomatitis virus (rVSVΔG-eGFP-NiVBD F/G) expressing NiV attachment (G) and fusion (F) glycoproteins. This recombinant virus serves as a valuable tool for investigating NiV entry mechanisms, cellular tropism, and immunogenicity. The virus was generated by replacing the VSV G protein with NiV F/G through reverse genetics, and protein incorporation was confirmed via immunofluorescence and electron microscopy. In vitro, the virus exhibited robust replication, characteristic cell tropism, and high viral titers in multiple cell lines. Neutralization assays showed that monoclonal antibodies HENV-26 and HENV-32 effectively neutralized the recombinant virus. Furthermore, immunization of golden hamsters with inactivated rVSVΔG-eGFP-NiVBD F/G induced potent neutralizing antibody responses, demonstrating its robust immunogenicity. These findings highlight rVSVΔG-eGFP-NiVBD F/G as an effective platform for NiV research and vaccine development. Full article
(This article belongs to the Section Animal Viruses)
Show Figures

Figure 1

15 pages, 770 KiB  
Review
Research Progress on the Gc Proteins of Akabane Virus
by Xiaolin Lan, Fang Liang, Gan Li, Weili Kong, Ruining Wang, Lin Wang, Mengmeng Zhao and Keshan Zhang
Vet. Sci. 2025, 12(8), 701; https://doi.org/10.3390/vetsci12080701 - 27 Jul 2025
Viewed by 227
Abstract
The Akabane virus (AKAV) is a significant member of the Orthobunyavirus genus, with its envelope glycoprotein Gc, focusing on its molecular structural features, immunoregulatory mechanisms, and application value in pathogen diagnosis and vaccine design. As a key structural protein of AKAV, Gc mediates [...] Read more.
The Akabane virus (AKAV) is a significant member of the Orthobunyavirus genus, with its envelope glycoprotein Gc, focusing on its molecular structural features, immunoregulatory mechanisms, and application value in pathogen diagnosis and vaccine design. As a key structural protein of AKAV, Gc mediates virus adsorption and neutralizing antibody recognition through the N-terminal highly variable region (HVR), while the C-terminal conserved region (CR) dominates the membrane fusion process, and its glycosylation modification has a significant regulatory effect on protein function. In clinical diagnostics, serological assays based on Gc proteins (e.g., ELISA, immunochromatographic test strips) have been standardized; in vaccine development, the neutralizing epitope of Gc proteins has become a core target for subunit vaccine design. Follow-up studies were deeply needed to analyze the structure-function interaction mechanism of Gc proteins to provide theoretical support for the construction of a new type of AKAV prevention and control system. Full article
Show Figures

Figure 1

16 pages, 2021 KiB  
Article
The Cytoplasmic Tail of Ovine Herpesvirus 2 Glycoprotein B Affects Cell Surface Expression and Is Required for Membrane Fusion
by Colleen M. Lynch, Maria K. Herndon, McKenna A. Hull, Daniela D. Moré, Katherine N. Baker, Cristina W. Cunha and Anthony V. Nicola
Viruses 2025, 17(7), 994; https://doi.org/10.3390/v17070994 - 16 Jul 2025
Viewed by 357
Abstract
Ovine herpesvirus 2 (OvHV-2) causes the fatal veterinary disease malignant catarrhal fever (MCF). Fusion is an essential step in the host cell entry of enveloped viruses and is an important target for vaccine development. OvHV-2 cannot be propagated in vitro, so a robust [...] Read more.
Ovine herpesvirus 2 (OvHV-2) causes the fatal veterinary disease malignant catarrhal fever (MCF). Fusion is an essential step in the host cell entry of enveloped viruses and is an important target for vaccine development. OvHV-2 cannot be propagated in vitro, so a robust virus-free cell–cell membrane fusion assay is necessary to elucidate its entry mechanism. OvHV-2 cell–cell fusion requires three conserved herpesviral envelope glycoproteins: gB, gH, and gL. OvHV-2 fusion activity is detectable but low. We hypothesize that enhancing the cell surface expression of gB, which is the core herpesviral fusogen, will increase cell–cell fusion. We generated C-terminal truncation mutants of gB and determined their cell surface expression, subcellular distribution, and fusion activity. Two mutants, including one that lacked the entire cytoplasmic tail domain, failed to function in the cell–cell fusion assay, despite wild-type levels of surface expression. This suggests that the OvHV-2 gB cytoplasmic tail is critical for fusion. A gB mutant truncated at amino acid 847 showed increased surface expression and fusion relative to the wild type. This suggests that the robust fusion activity of gB847 is the result of increased surface expression. gB847 may be used in place of wild-type gB in an improved, more robust OvHV-2 fusion assay. Full article
(This article belongs to the Section Animal Viruses)
Show Figures

Figure 1

20 pages, 6105 KiB  
Article
Potent Inhibition of Chikungunya Virus Entry by a Pyrazole–Benzene Derivative: A Computational Study Targeting the E1–E2 Glycoprotein Complex
by Md. Mohibur Rahman, Md. Belayet Hasan Limon, Tanvir Ahmed Saikat, Poulomi Saha, Abdul Hadi Nahid, Mohammad Mamun Alam and Mohammed Ziaur Rahman
Int. J. Mol. Sci. 2025, 26(13), 6480; https://doi.org/10.3390/ijms26136480 - 5 Jul 2025
Viewed by 564
Abstract
The Chikungunya virus (CHIKV) continues to pose a significant global health challenge due to the absence of effective antiviral treatments and limited vaccine availability. This study employed a comprehensive in silico workflow, incorporating high-throughput virtual screening, binding free-energy calculations, ADMET (absorption, distribution, metabolism, [...] Read more.
The Chikungunya virus (CHIKV) continues to pose a significant global health challenge due to the absence of effective antiviral treatments and limited vaccine availability. This study employed a comprehensive in silico workflow, incorporating high-throughput virtual screening, binding free-energy calculations, ADMET (absorption, distribution, metabolism, excretion, and toxicity) analysis, and 200 ns molecular dynamics (MD) simulations, to identify new inhibitors targeting the E1–E2 glycoprotein complex, crucial for CHIKV entry and membrane fusion. Four promising candidates were identified from a library of 20,000 compounds, with CID 136801451 showing the most potent binding (docking score: −10.227; ΔG_bind: −51.53 kcal/mol). The top four compounds exhibited favorable ADMET profiles, meeting nearly all criteria. MD simulations confirmed stable binding and strong interactions between CID 136801451 and the E1–E2 complex, evidenced by consistently low RMSD values. These findings highlight CID 136801451 as a promising CHIKV entry inhibitor, warranting further in vitro and in vivo evaluation to advance the development of effective anti-CHIKV therapeutics. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Graphical abstract

12 pages, 4737 KiB  
Communication
Myricetin Restricts the Syncytial Development Triggered by Nipah Virus Envelope Glycoproteins In Vitro
by Ananda Murali Rayapati, Chanda Chandrasekhar, Sudarsana Poojari and Bhadra Murthy Vemulapati
Viruses 2025, 17(6), 827; https://doi.org/10.3390/v17060827 - 7 Jun 2025
Viewed by 503
Abstract
Background and Objectives: Myricetin, a flavonoid compound, was demonstrated to effectively arrest the cell-to-cell fusion and syncytial development triggered by Nipah virus (NiV) fusion (F) and attachment (G) envelope glycoproteins in vitro involving two permissive mammalian cell lines. Methods: Time-of-addition assays were carried [...] Read more.
Background and Objectives: Myricetin, a flavonoid compound, was demonstrated to effectively arrest the cell-to-cell fusion and syncytial development triggered by Nipah virus (NiV) fusion (F) and attachment (G) envelope glycoproteins in vitro involving two permissive mammalian cell lines. Methods: Time-of-addition assays were carried out using codon-optimized NiV wild type (WT) F and G plasmids followed by a challenge with the addition of myricetin 1 h and 6 h post-transfection in HEK 293T and Vero cells. Results: Upon evaluating different myricetin concentrations, it was determined that a 100 μM concentration of myricetin effectively inhibited 64–80% of syncytia in HEK and Vero cells. Interpretation & Conclusions: In this study, we concluded that myricetin mitigated the syncytial development in HEK and Vero cell lines. Given the flavonoid attributes of myricetin which is widely present in fruits, vegetables, tea, and wine, it may be regarded as a phytonutrient and a safer antiviral alternative against Nipah virus infections. Due to the BSL-4 nature of the virus, further research involving live virus culture is necessary to confirm myricetin as a potential antiviral compound for the mitigation of pathological effects of NiV infections. Full article
(This article belongs to the Section General Virology)
Show Figures

Figure 1

17 pages, 1965 KiB  
Article
The Role of Long-Range Non-Specific Electrostatic Interactions in Inhibiting the Pre-Fusion Proteolytic Processing of the SARS-CoV-2 S Glycoprotein by Heparin
by Yi Du, Yang Yang, Son N. Nguyen and Igor A. Kaltashov
Biomolecules 2025, 15(6), 778; https://doi.org/10.3390/biom15060778 - 28 May 2025
Viewed by 488
Abstract
The proteolytic processing of the SARS-CoV-2 spike glycoprotein by host cell membrane-associated proteases is a key step in both the entry of the invading virus into the cell and the release of the newly generated viral particles from the infected cell. Because of [...] Read more.
The proteolytic processing of the SARS-CoV-2 spike glycoprotein by host cell membrane-associated proteases is a key step in both the entry of the invading virus into the cell and the release of the newly generated viral particles from the infected cell. Because of the critical importance of this step for the viral infectivity cycle, it has been a target of extensive efforts aimed at identifying highly specific protease inhibitors as potential antiviral agents. An alternative strategy to disrupt the pre-fusioviden processing of the SARS-CoV-2 S glycoprotein aims to protect the substrate rather than directly inhibit the proteases. In this work, we focused on furin, a serine protease located primarily in the Golgi apparatus, but also present on the cell membrane. Its cleavage site within the S glycoprotein is located within the stalk region of the latter and comprises an arginine-rich segment (SPRRARS), which fits the definition of the Cardin–Weintraub glycosaminoglycan recognition motif. Native mass spectrometry (MS) measurements confirmed the binding of a hexadecameric peptide representing the loop region at the S1/S2 interface and incorporating the furin cleavage site (FCS) to heparin fragments of various lengths, as well as unfractionated heparin (UFH), although at the physiological ionic strength, only UFH remains tightly bound to the FCS. The direct LC/MS monitoring of FCS digestion with furin revealed a significant impact of both heparin fragments and UFH on the proteolysis kinetics, although only the latter had IC50 values that could be considered physiologically relevant (0.6 ± 0.1 mg/mL). The results of this work highlight the importance of the long-range and relatively non-specific electrostatic interactions in modulating physiological and pathological processes and emphasize the multi-faceted role played by heparin in managing coronavirus infections. Full article
(This article belongs to the Special Issue Molecular Mechanism and Detection of SARS-CoV-2)
Show Figures

Figure 1

17 pages, 5361 KiB  
Article
Development and Potent Anti-Tumor Activity of a Fully Humanized Anti-TAG-72-IL-2 Fusion Protein for Therapy of Solid Tumors
by Eric Aniogo, Lindsay Williams, Teresa Hong, Patty Wong, Susanta K. Hui, Hemendra Ghimire, Erasmus K. Poku, David M. Colcher, Paul J. Yazaki, John E. Shively and Maciej Kujawski
Cancers 2025, 17(9), 1453; https://doi.org/10.3390/cancers17091453 - 26 Apr 2025
Viewed by 767
Abstract
Background/Objectives: Immunocytokines (ICKs) are antibody–cytokine fusion proteins that deliver cytokines directly to tumors to increase immune responses, which are otherwise absent or limited by the delivery of antibodies alone. Tumor-associated glycoprotein-72 (TAG72) is overexpressed in numerous solid tumors. Methods: An anti-TAG72-IL-2 fusion protein [...] Read more.
Background/Objectives: Immunocytokines (ICKs) are antibody–cytokine fusion proteins that deliver cytokines directly to tumors to increase immune responses, which are otherwise absent or limited by the delivery of antibodies alone. Tumor-associated glycoprotein-72 (TAG72) is overexpressed in numerous solid tumors. Methods: An anti-TAG72-IL-2 fusion protein was expressed in mammalian cells and tested in vitro for binding and bioactivity, and in vivo in two models. Results: In vitro studies showed high antigen specificity against TAG-72-positive tumor cell lines and IL-2 activity in CD25 (IL-2R)-positive reporter cells. To study the anti-tumor activity of huCC49-IL-2 in an immunocompetent model, the TAG-72 expression was established in murine mammary and colorectal cells by transfection with murine st6galnac-I gene (mSTn). Four daily doses of anti-TAG72-IL-2 monotherapy for TAG-72-expressing orthotopic murine mammary tumors in immunocompetent mice resulted in minimal whole-body toxicity and significant tumor growth reduction mediated by tumor infiltration of IFNγ+ CD8+ T cells. When mammary tumors were pretreated with image-guided fractionated radiation therapy (IGRT) followed by anti-TAG72-IL-2 therapy, an improved tumor growth inhibition was observed along with an increased tumor infiltration of IFNγ+ CD8+ T cells and a significant reduction in Foxp3+ CD4+ cells. Anti-TAG72-IL-2 monotherapy in TAG-72+ colorectal tumors led to a significant tumor reduction but also cures (4/7), with a rejection of rechallenges with both TAG-72-positive and -negative MC38 cells, thus demonstrating evidence of immune memory and antigen spreading. Conclusions: antiTAG-72-IL-2 therapy showed strong anti-tumor effects driven by activated CD8+ T cells making it a promising approach to the treatment of TAG-72+ tumors. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

15 pages, 1603 KiB  
Article
Quantifying Impact of HIV Receptor Surface Density Reveals Differences in Fusion Dynamics of HIV Strains
by Anthony Gerg and Hana M. Dobrovolny
Viruses 2025, 17(4), 583; https://doi.org/10.3390/v17040583 - 18 Apr 2025
Viewed by 493
Abstract
Human Immunodeficiency Virus (HIV) Type-1 has been studied heavily for decades, yet one area that is still poorly understood is the virus’ ability to cause cell–cell fusion. In HIV, the fusion process is mediated by viral surface glycoproteins that bind to CD4 cell [...] Read more.
Human Immunodeficiency Virus (HIV) Type-1 has been studied heavily for decades, yet one area that is still poorly understood is the virus’ ability to cause cell–cell fusion. In HIV, the fusion process is mediated by viral surface glycoproteins that bind to CD4 cell receptors. This virus-mediated cell fusion creates multi-nucleated cells called syncytia that can affect infection dynamics. Syncytia formation is often studied using a cell–cell fusion assay, in which donor cells expressing the viral surface protein fuse with acceptor cells expressing the cell receptor. A mathematical model capable of reproducing the dynamics of the cell–cell fusion assay was recently developed and can be used to quantify changes in syncytia formation. In this study, we use this mathematical model to quantify the changes in syncytia formation in HIV as the surface density of the glycoproteins is varied. We find that we need to modify the model to explicitly include a density-dependent syncytia formation rate that allows us to capture the dynamics of the cell–cell fusion assay as the density of the glycoproteins changes. With this modification, we find that cell–cell fusion of the HXB2 strain, which uses the CXCR4 coreceptor, shows a threshold-like behavior, while cell–cell fusion of the Sf162 strain, which uses the CCR5 co-receptor, shows a more gradual change as surface density decreases. Full article
Show Figures

Figure 1

9 pages, 2524 KiB  
Communication
Expression of Tailored α-N-Acetylglucosaminidase in Escherichia coli for Synthesizing Mannose-6-Phosphate on N-Linked Oligosaccharides of Lysosomal Enzymes
by Yunsong Cao and Wei Wang
Bioengineering 2025, 12(4), 425; https://doi.org/10.3390/bioengineering12040425 - 17 Apr 2025
Viewed by 473
Abstract
Lysosomal enzymes are synthesized as N-glycosylated glycoproteins with mannose-6-phosphate (M6P) moieties, which are responsible for their binding to M6P receptors and transporting to the lysosome. In the M6P biosynthetic pathway, a Man8GlcNAc2 glycoform is converted to M6P groups through two [...] Read more.
Lysosomal enzymes are synthesized as N-glycosylated glycoproteins with mannose-6-phosphate (M6P) moieties, which are responsible for their binding to M6P receptors and transporting to the lysosome. In the M6P biosynthetic pathway, a Man8GlcNAc2 glycoform is converted to M6P groups through two consecutive enzymatic reactions, including N-acetylglucosamine (GlcNAc)-1-phosphotransferase (GNPT), transferring GlcNAc-1-phosphate from UDP-GlcNAc to the C6 hydroxyl groups of mannose residues, and then, removal of the covering GlcNAc moiety from the GlcNAc-P-mannose phosphodiester was carried out using an α-N-acetylglucosaminidase (referred to as ‘uncovering enzyme’, UCE) in the trans-Golgi network (TGN). Here, we expressed differently tailored versions of the UCE, including four truncated variants, in Escherichia coli. The four variants with the signal peptide, transmembrane domain, propiece and cytoplasmic tail truncated, respectively, were purified by affinity chromatography, and their enzymatic activities were assayed using a UDP-Glo kit. By fusing a maltose-binding protein (MBP) in the N-terminus of the UCE variants, the fusion proteins could be soluble when expressed in E. coli. The highest concentration of the purified enzyme was 80.5 mg/L of fermentation broth. Furthermore, the UCE with the core catalytic domain exhibited the highest uncovering activity. Full article
(This article belongs to the Section Biochemical Engineering)
Show Figures

Graphical abstract

18 pages, 6369 KiB  
Review
Progress on Respiratory Syncytial Virus Vaccine Development and Evaluation Methods
by Lie Deng, Hongjie Cao, Guichang Li, Kaiwen Zhou, Zihan Fu, Jiaying Zhong, Zhongfang Wang and Xiaoyun Yang
Vaccines 2025, 13(3), 304; https://doi.org/10.3390/vaccines13030304 - 12 Mar 2025
Cited by 1 | Viewed by 2438
Abstract
Respiratory syncytial virus (RSV) remains a significant global health threat, especially to infants, the elderly, and immunocompromised individuals. This review comprehensively explores the progress in RSV vaccine development, the immune evaluation methods, and immunological surrogate. The RSV fusion (F) protein, a primary target [...] Read more.
Respiratory syncytial virus (RSV) remains a significant global health threat, especially to infants, the elderly, and immunocompromised individuals. This review comprehensively explores the progress in RSV vaccine development, the immune evaluation methods, and immunological surrogate. The RSV fusion (F) protein, a primary target for vaccine development, has been engineered in prefusion conformation to elicit potent neutralizing antibodies, while the attachment (G) glycoprotein and other immunogens are also being explored to broaden immune responses. Advances in diverse vaccine platforms, ranging from live attenuated and protein subunit vaccines to cutting-edge mRNA- and nanoparticle-based formulations, highlight the field’s progress, yet challenges in balancing safety, immunogenicity, and durability persist. Central to these efforts is the identification and validation of immunological surrogates, which may serve as critical benchmarks for vaccine efficacy. Neutralizing antibody titers, multifunctional T cell responses, and B cell memory have emerged as key correlates of protection. However, the feasibility of these surrogates depends on their ability to predict clinical outcomes across diverse populations and settings. While neutralizing antibodies block the virus directly, T cell responses are essential for clearing infected cells and preventing severe disease, and B cell memory ensures long-term immunity. Integrating these immunological markers into a cohesive framework requires standardized assays, robust clinical validation, and an in-depth understanding of RSV-induced immune response. Full article
(This article belongs to the Topic Advances in Vaccines and Antimicrobial Therapy)
Show Figures

Figure 1

14 pages, 2996 KiB  
Article
Structures of HCoV-OC43 HR1 Domain in Complex with Cognate HR2 or Analogue EK1 Peptide
by Xiuxiu He, Huanzhen Liu, Guang Yang and Lei Yan
Viruses 2025, 17(3), 343; https://doi.org/10.3390/v17030343 - 28 Feb 2025
Viewed by 625
Abstract
Human coronavirus OC43 (HCoV-OC43) is usually associated with common colds, but also related to severe disease in the frail. Its envelope glycoproteins spike (S) is responsible for host-cell attachment and membrane fusion. To understand the molecular basis of membrane fusion of HCoV-OC43, we [...] Read more.
Human coronavirus OC43 (HCoV-OC43) is usually associated with common colds, but also related to severe disease in the frail. Its envelope glycoproteins spike (S) is responsible for host-cell attachment and membrane fusion. To understand the molecular basis of membrane fusion of HCoV-OC43, we solved the 3.34 Å crystal structure of the post-fusion state formed by two heptad repeat domains (HR1P and HR2P) of OC43-S. This fusion core comprises a parallel trimeric coiled coil of three HR1 helices with 61 Å at length, around which three HR2 helices are entwined in an antiparallel manner, as anticipated. Moreover, a pan-CoV fusion inhibitor EK1 derived from OC43-HR2P was also crystalized with OC43-HR1P in the resolution of 2.71 Å. Parallel comparisons rationalize the design of EK1, maintaining various hydrophobic and charged or hydrophilic interactions formed in the initial fusion core to stabilize the overall conformation. Together, our results not only reveal the critical intrahelical and interhelical interactions underlying the mechanism of action of OC43-S fusion, but also help our understanding on the mechanism of HCoV-OC43 inhibition by analogue HR2 mimic peptide. Full article
(This article belongs to the Special Issue Coronaviruses Pathogenesis, Immunity, and Antivirals (2nd Edition))
Show Figures

Figure 1

22 pages, 4211 KiB  
Article
Flunarizine as a Candidate for Drug Repurposing Against Human Pathogenic Mammarenaviruses
by Chukwudi A. Ofodile, Ikemefuna C. Uzochukwu, Fortunatus C. Ezebuo, InnocentMary Ejiofor, Mercy Adebola, Innocent Okpoli, Beatrice Cubitt, Haydar Witwit, Chetachi B. Okwuanaso, Ngozi Onyemelukwe and Juan Carlos de la Torre
Viruses 2025, 17(1), 117; https://doi.org/10.3390/v17010117 - 16 Jan 2025
Viewed by 1208
Abstract
Lassa fever (LF), a viral hemorrhagic fever disease with a case fatality rate that can be over 20% among hospitalized LF patients, is endemic to many West African countries. Currently, no vaccines or therapies are specifically licensed to prevent or treat LF, hence [...] Read more.
Lassa fever (LF), a viral hemorrhagic fever disease with a case fatality rate that can be over 20% among hospitalized LF patients, is endemic to many West African countries. Currently, no vaccines or therapies are specifically licensed to prevent or treat LF, hence the significance of developing therapeutics against the mammarenavirus Lassa virus (LASV), the causative agent of LF. We used in silico docking approaches to investigate the binding affinities of 2015 existing drugs to LASV proteins known to play critical roles in the formation and activity of the virus ribonucleoprotein complex (vRNP) responsible for directing replication and transcription of the viral genome. Validation of docking protocols were achieved with reference inhibitors of the respective targets. Our in silico docking screen identified five drugs (dexamethasone, tadalafil, mefloquine, ergocalciferol, and flunarizine) with strong predicted binding affinity to LASV proteins involved in the formation of the vRNP. We used cell-based functional assays to evaluate the antiviral activity of the five selected drugs. We found that flunarizine, a calcium-entry blocker, inhibited the vRNP activity of LASV and LCMV and virus surface glycoprotein fusion activity required for mammarenavirus cell entry. Consistently with these findings, flunarizine significantly reduced peak titers of LCMV in a multi-step growth kinetics assay in human A549 cells. Flunarizine is being used in several countries worldwide to treat vertigo and migraine, supporting the interest in exploring its repurposing as a candidate drug to treat LASV infections. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

15 pages, 8637 KiB  
Article
SSL5-AnxA5 Fusion Protein Constructed Based on Human Atherosclerotic Plaque scRNA-Seq Data Preventing the Binding of Apoptotic Endothelial Cells, Platelets, and Inflammatory Cells
by Yifei Zhao, Xingyu He, Teng Hu, Tianli Xia, Fangyang Huang, Changming Li, Yiming Li, Fei Chen, Mao Chen, Jun Ma and Yong Peng
Biomedicines 2025, 13(1), 8; https://doi.org/10.3390/biomedicines13010008 - 24 Dec 2024
Viewed by 982
Abstract
Background and aims: Coronary obstruction following plaque rupture is a critical pathophysiological change in the progression of stable angina (SAP) to acute coronary syndrome (ACS). The accumulation of platelets and various inflammatory cells on apoptotic endothelial cells is a key factor in arterial [...] Read more.
Background and aims: Coronary obstruction following plaque rupture is a critical pathophysiological change in the progression of stable angina (SAP) to acute coronary syndrome (ACS). The accumulation of platelets and various inflammatory cells on apoptotic endothelial cells is a key factor in arterial obstruction after plaque rupture. Through single-cell sequencing analysis (scRNA-seq) of plaques from SAP and ACS patients, we identified significant changes in the annexin V and P-selectin glycoprotein ligand 1 pathways. Staphylococcal superantigen-like 5 (SSL5) is an optimal antagonist P-selectin glycoprotein ligand 1 (PSGL1), while annexin V (AnxA5) can precisely detect dead cells in vivo. We constructed the SSL5-AnxA5 fusion protein and observed its role in preventing the interaction between apoptotic endothelial cells, platelets, and inflammatory cells. Methods: The scRNA-seq data were extracted from the Gene Expression Omnibus (GEO) database. Single-cell transcriptome analysis results and cell–cell communication were analyzed to identify the ACS and SAP cell clusters and elucidate the intercellular communication differences. Then, we constructed and verified a fusion protein comprising SSL5 and AnxA5 domains via polymerase chain reaction (PCR) and Western blot. The binding capacity of the fusion protein to P-selectin and apoptotic cells was evaluated by flow cytometry and AnxA5-FITC apoptosis detection kit, respectively. Furthermore, co-incubation and immunofluorescence allowed us to describe the mediation effect of it between inflammatory cells and endothelial cells or activated platelets. Results: Our analysis of the scRNA-seq data showed that SELPLG (PSGL1 gene) and ANNEXIN had higher information flowing in ACS compared to SAP. The SELPLG signaling pathway network demonstrated a higher number of interactions in ACS, while the ANNEXIN signaling pathway network revealed stronger signaling from macrophages toward monocytes in ACS compared to SAP. Competition binding experiments with P-selectin showed that SSL5-AnxA5 induced a decrease in the affinity of PSGL1. SSL5-AnxA5 effectively inhibited the combination of endothelial cells with inflammatory cells and the interaction of activated platelets with inflammatory cells. Additionally, this fusion protein exhibited remarkable capability in binding to apoptotic cells. Conclusions: The bifunctional protein SSL5-AnxA5 exhibits promising potential as a protective agent against local inflammation in arterial tissues, making it an excellent candidate for PSGL1-related therapeutic interventions. Full article
(This article belongs to the Special Issue Angiogenesis and Related Disorders)
Show Figures

Figure 1

19 pages, 1776 KiB  
Review
Decoding the Genes Orchestrating Egg and Sperm Fusion Reactions and Their Roles in Fertility
by Ranjha Khan, Muhammad Azhar and Muhammad Umair
Biomedicines 2024, 12(12), 2850; https://doi.org/10.3390/biomedicines12122850 - 15 Dec 2024
Viewed by 2627
Abstract
Mammalian fertilization is a complex and highly regulated process that has garnered significant attention, particularly with advancements in assisted reproductive technologies such as in vitro fertilization (IVF). The fusion of egg and sperm involves a sequence of molecular and cellular events, including capacitation, [...] Read more.
Mammalian fertilization is a complex and highly regulated process that has garnered significant attention, particularly with advancements in assisted reproductive technologies such as in vitro fertilization (IVF). The fusion of egg and sperm involves a sequence of molecular and cellular events, including capacitation, the acrosome reaction, adhesion, and membrane fusion. Critical genetic factors, such as IZUMO1, JUNO (also known as FOLR4), CD9, and several others, have been identified as essential mediators in sperm–egg recognition and membrane fusion. Additionally, glycoproteins such as ZP3 within the zona pellucida are crucial for sperm binding and triggering the acrosome reaction. Recent gene-editing technologies, such as CRISPR/Cas9 and conditional knockout models, have facilitated the functional annotation of genes such as SPAM1 and ADAM family members, further elucidating their roles in capacitation and adhesion. Furthermore, the integration of CRISPR-Cas9 with omics technologies, including transcriptomics, proteomics, and lipidomics, has unlocked new avenues for identifying previously unknown genetic players and pathways involved in fertilization. For instance, transcriptomics can uncover gene expression profiles during gamete maturation, while proteomics identifies key protein interactions critical for processes such as capacitation and the acrosome reaction. Lipidomics adds another dimension by revealing how membrane composition influences gamete fusion. Together, these tools enable the discovery of novel genes, pathways, and molecular mechanisms involved in fertility, providing insights that were previously unattainable. These approaches not only deepen our molecular understanding of fertility mechanisms but also hold promise for refining diagnostic tools and therapeutic interventions for infertility. This review summarizes the current molecular insights into genes orchestrating fertilization and highlights cutting-edge methodologies that propel the field toward novel discoveries. By integrating these findings, this review aims to provide valuable knowledge for clinicians, researchers, and technologists in the field of reproductive biology and assisted reproductive technologies. Full article
(This article belongs to the Special Issue Molecular and Genetic Bases of Infertility)
Show Figures

Figure 1

25 pages, 6325 KiB  
Article
An In Silico Approach to Discover Efficient Natural Inhibitors to Tie Up Epstein–Barr Virus Infection
by Ayan Das, Mumtaza Mumu, Tanjilur Rahman, Md Abu Sayeed, Md Mazharul Islam, John I. Alawneh and Mohammad Mahmudul Hassan
Pathogens 2024, 13(11), 928; https://doi.org/10.3390/pathogens13110928 - 25 Oct 2024
Viewed by 2431
Abstract
Epstein–Barr virus (EBV), also known as human herpesvirus 4, is a member of the herpes virus family. EBV is a widespread virus and causes infectious mononucleosis, which manifests with symptoms such as fever, fatigue, lymphadenopathy, splenomegaly, and hepatomegaly. Additionally, EBV is associated with [...] Read more.
Epstein–Barr virus (EBV), also known as human herpesvirus 4, is a member of the herpes virus family. EBV is a widespread virus and causes infectious mononucleosis, which manifests with symptoms such as fever, fatigue, lymphadenopathy, splenomegaly, and hepatomegaly. Additionally, EBV is associated with different lymphocyte-associated non-malignant, premalignant, and malignant diseases. So far, no effective treatment or therapeutic drug is known for EBV-induced infections and diseases. This study investigated natural compounds that inhibit EBV glycoprotein L (gL) and block EBV fusion in host cells. We utilised computational approaches, including molecular docking, in silico ADMET analysis, and molecular dynamics simulation. We docked 628 natural compounds against gL and identified the four best compounds based on binding scores and pharmacokinetic properties. These four compounds, with PubChem CIDs 4835509 (CHx-HHPD-Ac), 2870247 (Cyh-GlcNAc), 21206004 (Hep-HHPD-Ac), and 51066638 (Und-GlcNAc), showed several interactions with EBV gL. However, molecular dynamics simulations indicated that the protein–ligand complexes of CID: 4835509 (CHx-HHPD-Ac) and CID: 2870247 (Cyh-GlcNAc) are more stable than those of the other two compounds. Therefore, CIDs 4835509 and 2870247 (Cyh-GlcNAc) may be potent natural inhibitors of EBV infection. These findings can open a new way for effective drug design against EBV and its associated infections and diseases. Full article
Show Figures

Figure 1

Back to TopTop