Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.913
Filter
1.
BMC Vet Res ; 20(1): 288, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961481

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) mainly causes acute and severe porcine epidemic diarrhea (PED), and is highly fatal in neonatal piglets. No reliable therapeutics against the infection exist, which poses a major global health issue for piglets. Luteolin is a flavonoid with anti-viral activity toward several viruses. RESULTS: We evaluated anti-viral effects of luteolin in PEDV-infected Vero and IPEC-J2 cells, and identified IC50 values of 23.87 µM and 68.5 µM, respectively. And found PEDV internalization, replication and release were significantly reduced upon luteolin treatment. As luteolin could bind to human ACE2 and SARS-CoV-2 main protease (Mpro) to contribute viral entry, we first identified that luteolin shares the same core binding site on pACE2 with PEDV-S by molecular docking and exhibited positive pACE2 binding with an affinity constant of 71.6 µM at dose-dependent increases by surface plasmon resonance (SPR) assay. However, pACE2 was incapable of binding to PEDV-S1. Therefore, luteolin inhibited PEDV internalization independent of PEDV-S binding to pACE2. Moreover, luteolin was firmly embedded in the groove of active pocket of Mpro in a three-dimensional docking model, and fluorescence resonance energy transfer (FRET) assays confirmed that luteolin inhibited PEDV Mpro activity. In addition, we also observed PEDV-induced pro-inflammatory cytokine inhibition and Nrf2-induced HO-1 expression. Finally, a drug resistant mutant was isolated after 10 cell culture passages concomitant with increasing luteolin concentrations, with reduced PEDV susceptibility to luteolin identified at passage 10. CONCLUSIONS: Our results push forward that anti-PEDV mechanisms and resistant-PEDV properties for luteolin, which may be used to combat PED.


Subject(s)
Antiviral Agents , Luteolin , Porcine epidemic diarrhea virus , Luteolin/pharmacology , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Vero Cells , Swine , Molecular Docking Simulation , Virus Internalization/drug effects , Virus Replication/drug effects , Cell Line , Computer Simulation , Swine Diseases/virology , Swine Diseases/drug therapy
2.
Nat Commun ; 15(1): 5606, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961055

ABSTRACT

Viral mutations frequently outpace technologies used to detect harmful variants. Given the continual emergence of SARS-CoV-2 variants, platforms that can identify the presence of a virus and its propensity for infection are needed. Our electronic biomembrane sensing platform recreates distinct SARS-CoV-2 host cell entry pathways and reports the progression of entry as electrical signals. We focus on two necessary entry processes mediated by the viral Spike protein: virus binding and membrane fusion, which can be distinguished electrically. We find that closely related variants of concern exhibit distinct fusion signatures that correlate with trends in cell-based infectivity assays, allowing us to report quantitative differences in their fusion characteristics and hence their infectivity potentials. We use SARS-CoV-2 as our prototype, but we anticipate that this platform can extend to other enveloped viruses and cell lines to quantifiably assess virus entry.


Subject(s)
COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Humans , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , COVID-19/virology , Membrane Fusion , Cell-Free System , Mutation , Virus Attachment
4.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38928089

ABSTRACT

SARS-CoV-2 S-protein-mediated fusion is thought to involve the interaction of the membrane-distal or N-terminal heptad repeat (NHR) ("HR1") of the cleaved S2 segment of the protein and the membrane-proximal or C-terminal heptad repeat (CHR) ("HR2") regions of the protein. We examined the fusion inhibitory activity of a PEGylated HR2-derived peptide and its palmitoylated derivative using a pseudovirus infection assay. The latter peptide caused a 76% reduction in fusion activity at 10 µM. Our results suggest that small variations in peptide derivatization and differences in the membrane composition of pseudovirus preparations may affect the inhibitory potency of HR2-derived peptides. We suggest that future studies on the inhibition of infectivity of SARS-CoV-2 in both in vitro and in vivo systems consider the need for higher concentrations of peptide inhibitors.


Subject(s)
Peptides , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Humans , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Peptides/pharmacology , Peptides/chemistry , Palmitic Acid/pharmacology , Palmitic Acid/chemistry , Virus Internalization/drug effects , COVID-19/virology , COVID-19/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry
5.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928216

ABSTRACT

The pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global health concern. Cell entry of SARS-CoV-2 depends on viral spike (S) proteins binding to cellular receptors (ACE2) and their subsequent priming by host cell proteases (TMPRSS2). Assessing effects of viral-induced host response factors and determining which cells are used by SARS-CoV-2 for entry might provide insights into viral transmission, add clarity to the virus' pathogenesis, and possibly reveal therapeutic targets. Mast cells (MCs) are ubiquitously expressed tissue cells that act as immune sentinels given their ability to react specifically to pathogens at environmental interfaces, such as in the lung. Several lines of evidence suggest a critical role for MCs in SARS-CoV-2 infections based on patients' mediator profiles, especially the "cytokine storm" responsible for most morbidity and mortality. In this pilot study, we demonstrated that human lung MCs (n = 3 donors) are a source of renin and that they upregulate the membrane receptor for SARS-CoV-2 (ACE2) as well as the protease required for cellular entry (TMPRSS2) under certain conditions. We hypothesized that infection of human MCs with SARS-CoV-2 may be a heretofore-unrecognized mechanism of viral pathogenesis, and further studies are required to assess this question.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Lung , Mast Cells , SARS-CoV-2 , Serine Endopeptidases , Humans , Mast Cells/virology , Mast Cells/immunology , Mast Cells/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/immunology , SARS-CoV-2/physiology , COVID-19/virology , COVID-19/immunology , COVID-19/pathology , Angiotensin-Converting Enzyme 2/metabolism , Lung/virology , Lung/pathology , Lung/immunology , Serine Endopeptidases/metabolism , Virus Internalization , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/immunology
6.
Virulence ; 15(1): 2367671, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38910312

ABSTRACT

Viral diseases are among the main threats to public health. Understanding the factors affecting viral invasion is important for antiviral research. Until now, it was known that most viruses have very low plaque-forming unit (PFU)-to-particle ratios. However, further investigation is required to determine the underlying factors. Here, using quantitative single-particle analysis methods, the invasion of Semliki Forest virus (SFV), Japanese encephalitis virus (JEV), and influenza A virus (IAV) containing attachment to the cell surface, entry into the cell, transport towards the cell interior, and fusion with endosomes to release nucleocapsids were quantitatively analysed in parallel. It was found that for SFV with an PFU-to-particle ratio of approximately 1:2, an entry efficiency of approximately 31% limited infection. For JEV, whose PFU-to-particle ratio was approximately 1:310, an attachment efficiency of approximately 27% and an entry efficiency of 10% were the main factors limiting its infection. Meanwhile, for IAV with PFU-to-particle ratios of 1:8100, 5% attachment efficiency, 9% entry efficiency, and 53% fusion efficiency significantly limited its infection. These results suggest that viruses with different infectivities have different limited steps in the invasion process. Moreover, there are significant differences in attachment efficiencies among viruses, emphasizing the pivotal role of attachment in viral invasion. The influence of the virus purification method on virus invasion was also investigated. This study, for the first time, reports the efficiencies of different stages of virus invasion, leading to a better understanding of virus invasion and providing a protocol to quantitatively analyse the virus invasion efficiency.


Subject(s)
Influenza A virus , Semliki forest virus , Virus Internalization , Influenza A virus/physiology , Animals , Semliki forest virus/physiology , Humans , Encephalitis Virus, Japanese/physiology , Cell Line , Virus Attachment , Endosomes/virology
7.
Chem Biol Drug Des ; 103(6): e14566, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38858134

ABSTRACT

The severe acute respiratory syndrome coronavirus (SARS-CoV-2) pandemic has triggered a significant impact on global public health security, it is urgent to develop effective antiviral drugs. Previous studies have found that binding to ACE2 is a key step in the invasion of SARS-CoV-2 into host cells, so virus invasion can be inhibited by blocking ACE2, but there are few reports on this kind of specific inhibitor. Our previous study found that Harringtonine (HT) can inhibit the entry of SARS-CoV-2 spike pseudovirus into ACE2h cells, but its relatively high cytotoxicity limits its further development. Amino acid modification of the active components can increase their solubility and reduce their cytotoxicity. Therefore, in this study, seven new derivatives were synthesized by amino acid modification of its core structure Cephalotaxine. The target compounds were evaluated by cell viability assay and the SARS-CoV-2 spike pseudovirus entry assay. Compound CET-1 significantly inhibited the entry of pseudovirus into ACE2h cells and showed less cytotoxicity than HT. Molecular docking results showed that CET-1 could bind TYR83, an important residue of ACE2, just like HT. In conclusion, our study provided a novel compound with more potential activity and lower toxicity than HT on inhibiting the SARS-CoV-2 spike pseudovirus infection, which makes it possible to be a lead compound as an antiviral drug in the future.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antiviral Agents , COVID-19 Drug Treatment , Homoharringtonine , Molecular Docking Simulation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Amino Acids/chemistry , Amino Acids/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Cell Survival/drug effects , COVID-19/virology , Homoharringtonine/pharmacology , Homoharringtonine/chemistry , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/chemistry , Virus Internalization/drug effects , Harringtonines/chemistry , Harringtonines/pharmacology
8.
Microbiol Spectr ; 12(7): e0422023, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38864599

ABSTRACT

The emergence of the COVID-19 pandemic prompted an increased interest in seasonal human coronaviruses. OC43, 229E, NL63, and HKU1 are endemic seasonal coronaviruses that cause the common cold and are associated with generally mild respiratory symptoms. In this study, we identified cell lines that exhibited cytopathic effects (CPE) upon infection by three of these coronaviruses and characterized their viral replication kinetics and the effect of infection on host surface receptor expression. We found that NL63 produced CPE in LLC-MK2 cells, while OC43 produced CPE in MRC-5, HCT-8, and WI-38 cell lines, while 229E produced CPE in MRC-5 and WI-38 by day 3 post-infection. We observed a sharp increase in nucleocapsid and spike viral RNA (vRNA) from day 3 to day 5 post-infection for all viruses; however, the abundance and the proportion of vRNA copies measured in the supernatants and cell lysates of infected cells varied considerably depending on the virus-host cell pair. Importantly, we observed modulation of coronavirus entry and attachment receptors upon infection. Infection with 229E and OC43 led to a downregulation of CD13 and GD3, respectively. In contrast, infection with NL63 and OC43 leads to an increase in ACE2 expression. Attempts to block entry of NL63 using either soluble ACE2 or anti-ACE2 monoclonal antibodies demonstrated the potential of these strategies to greatly reduce infection. Overall, our results enable a better understanding of seasonal coronaviruses infection kinetics in permissive cell lines and reveal entry receptor modulation that may have implications in facilitating co-infections with multiple coronaviruses in humans.IMPORTANCESeasonal human coronavirus is an important cause of the common cold associated with generally mild upper respiratory tract infections that can result in respiratory complications for some individuals. There are no vaccines available for these viruses, with only limited antiviral therapeutic options to treat the most severe cases. A better understanding of how these viruses interact with host cells is essential to identify new strategies to prevent infection-related complications. By analyzing viral replication kinetics in different permissive cell lines, we find that cell-dependent host factors influence how viral genes are expressed and virus particles released. We also analyzed entry receptor expression on infected cells and found that these can be up- or down-modulated depending on the infecting coronavirus. Our findings raise concerns over the possibility of infection enhancement upon co-infection by some coronaviruses, which may facilitate genetic recombination and the emergence of new variants and strains.


Subject(s)
Coronavirus 229E, Human , Coronavirus NL63, Human , Coronavirus OC43, Human , Virus Internalization , Virus Replication , Humans , Coronavirus NL63, Human/physiology , Coronavirus NL63, Human/genetics , Coronavirus 229E, Human/physiology , Coronavirus 229E, Human/genetics , Coronavirus OC43, Human/physiology , Coronavirus OC43, Human/genetics , Cell Line , Seasons , Kinetics , Receptors, Virus/metabolism , Receptors, Virus/genetics , Common Cold/virology , Common Cold/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Viral/metabolism , RNA, Viral/genetics , Animals , COVID-19/virology , COVID-19/metabolism , Coronavirus/physiology , Coronavirus/genetics
9.
Adv Virus Res ; 119: 1-38, 2024.
Article in English | MEDLINE | ID: mdl-38897707

ABSTRACT

The ubiquitination process is a reversible posttranslational modification involved in many essential cellular functions, such as innate immunity, cell signaling, trafficking, protein stability, and protein degradation. Viruses can use the ubiquitin system to efficiently enter host cells, replicate and evade host immunity, ultimately enhancing viral pathogenesis. Emerging evidence indicates that enveloped viruses can carry free (unanchored) ubiquitin or covalently ubiquitinated viral structural proteins that can increase the efficiency of viral entry into host cells. Furthermore, viruses continuously evolve and adapt to take advantage of the host ubiquitin machinery, highlighting its importance during virus infection. This review discusses the battle between viruses and hosts, focusing on how viruses hijack the ubiquitination process at different steps of the replication cycle, with a specific emphasis on viral entry. We discuss how ubiquitination of viral proteins may affect tropism and explore emerging therapeutics strategies targeting the ubiquitin system for antiviral drug discovery.


Subject(s)
Ubiquitination , Virus Internalization , Virus Replication , Humans , Ubiquitin/metabolism , Viruses/metabolism , Host-Pathogen Interactions , Viral Proteins/metabolism , Viral Proteins/genetics , Virus Diseases/virology , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Protein Processing, Post-Translational
10.
Nat Microbiol ; 9(7): 1764-1777, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849624

ABSTRACT

Influenza virus infection is initiated by the attachment of the viral haemagglutinin (HA) protein to sialic acid receptors on the host cell surface. Most virus particles enter cells through clathrin-mediated endocytosis (CME). However, it is unclear how viral binding signals are transmitted through the plasma membrane triggering CME. Here we found that metabotropic glutamate receptor subtype 2 (mGluR2) and potassium calcium-activated channel subfamily M alpha 1 (KCa1.1) are involved in the initiation and completion of CME of influenza virus using an siRNA screen approach. Influenza virus HA directly interacted with mGluR2 and used it as an endocytic receptor to initiate CME. mGluR2 interacted and activated KCa1.1, leading to polymerization of F-actin, maturation of clathrin-coated pits and completion of the CME of influenza virus. Importantly, mGluR2-knockout mice were significantly more resistant to different influenza subtypes than the wild type. Therefore, blocking HA and mGluR2 interaction could be a promising host-directed antiviral strategy.


Subject(s)
Endocytosis , Mice, Knockout , Receptors, Metabotropic Glutamate , Animals , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/genetics , Mice , Humans , Virus Internalization , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Clathrin/metabolism , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/metabolism , HEK293 Cells , Actins/metabolism , Dogs , Madin Darby Canine Kidney Cells , Receptors, Virus/metabolism , Receptors, Virus/genetics , Influenza, Human/virology , Influenza, Human/metabolism , Orthomyxoviridae/physiology , Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism
11.
Front Cell Infect Microbiol ; 14: 1388360, 2024.
Article in English | MEDLINE | ID: mdl-38841111

ABSTRACT

Background: Alphaviruses are a diverse group of pathogens that have garnered considerable attention due to their impact on human health. By investigating alphavirus receptors, researchers can elucidate viral entry mechanisms and gain important clues for the prevention and treatment of viral diseases. This study presents an in-depth analysis of the research progress made in the field of alphavirus receptors through bibliometric analysis. Methods: This study encompasses various aspects, including historical development, annual publication trends, author and cited-author analysis, institutional affiliations, global distribution of research contributions, reference analysis with strongest citation bursts, keyword analysis, and a detailed exploration of recent discoveries in alphavirus receptor research. Results: The results of this bibliometric analysis highlight key milestones in alphavirus receptor research, demonstrating the progression of knowledge in this field over time. Additionally, the analysis reveals current research hotspots and identifies emerging frontiers, which can guide future investigations and inspire novel therapeutic strategies. Conclusion: This study provides an overview of the state of the art in alphavirus receptor research, consolidating the existing knowledge and paving the way for further advancements. By shedding light on the significant developments and emerging areas of interest, this study serves as a valuable resource for researchers, clinicians, and policymakers engaged in combating alphavirus infections and improving public health.


Subject(s)
Alphavirus , Bibliometrics , Humans , Receptors, Virus/metabolism , Animals , Virus Internalization , Alphavirus Infections/virology , Biomedical Research/trends
12.
Molecules ; 29(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38893512

ABSTRACT

COVID-19 continues to spread around the world. This is mainly because new variants of the SARS-CoV-2 virus emerge due to genomic mutations, evade the immune system and result in the effectiveness of current therapeutics being reduced. We previously established a series of detection platforms, comprising computational docking analysis, S-protein-based ELISA, pseudovirus entry, and 3CL protease activity assays, which allow us to screen a large library of phytochemicals from natural products and to determine their potential in blocking the entry of SARS-CoV-2. In this new screen, rutaecarpine (an alkaloid from Evodia rutaecarpa) was identified as exhibiting anti-SARS-CoV-2 activity. Therefore, we conducted multiple rounds of structure-activity-relationship (SAR) studies around this phytochemical and generated several rutaecarpine analogs that were subjected to in vitro evaluations. Among these derivatives, RU-75 and RU-184 displayed remarkable inhibitory activity when tested in the 3CL protease assay, S-protein-based ELISA, and pseudovirus entry assay (for both wild-type and omicron variants), and they attenuated the inflammatory response induced by SARS-CoV-2. Interestingly, RU-75 and RU-184 both appeared to be more potent than rutaecarpine itself, and this suggests that they might be considered as lead candidates for future pharmacological elaboration.


Subject(s)
Antiviral Agents , Drug Design , Indole Alkaloids , Molecular Docking Simulation , Quinazolines , SARS-CoV-2 , Indole Alkaloids/pharmacology , Indole Alkaloids/chemistry , SARS-CoV-2/drug effects , Quinazolines/pharmacology , Quinazolines/chemistry , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Structure-Activity Relationship , COVID-19 Drug Treatment , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Virus Internalization/drug effects , Quinazolinones
13.
Front Cell Infect Microbiol ; 14: 1335189, 2024.
Article in English | MEDLINE | ID: mdl-38895735

ABSTRACT

Background: Chikungunya virus (CHIKV), which causes chikungunya fever, is an arbovirus of public health concern with no approved antiviral therapies. A significant proportion of patients develop chronic arthritis after an infection. Zinc and magnesium salts help the immune system respond effectively against viral infections. This study explored the antiviral potential of zinc sulphate, zinc acetate, and magnesium sulphate against CHIKV infection. Methods: The highest non-toxic concentration of the salts (100 µM) was used to assess the prophylactic, virucidal, and therapeutic anti-CHIKV activities. Dose-dependent antiviral effects were investigated to find out the 50% inhibitory concentration of the salts. Entry bypass assay was conducted to find out whether the salts affect virus entry or post entry stages. Virus output in all these experiments was estimated using a focus-forming unit assay, real-time RT-PCR, and immunofluorescence assay. Results: Different time- and temperature-dependent assays revealed the therapeutic antiviral activity of zinc and magnesium salts against CHIKV. A minimum exposure of 4 hours and treatment initiation within 1 to 2 hours of infection are required for inhibition of CHIKV. Entry assays revealed that zinc salt affected virus-entry. Entry bypass assays suggested that both salts affected post-entry stages of CHIKV. In infected C57BL6 mice orally fed with zinc and magnesium salts, a reduction in viral RNA copy number was observed. Conclusion: The study results suggest zinc salts exert anti-CHIKV activity at entry and post entry stages of the virus life cycle, while magnesium salt affect CHIKV at post entry stages. Overall, the study highlights the significant antiviral potential of zinc sulphate, zinc acetate, and magnesium sulphate against CHIKV, which can be exploited in designing potential therapeutic strategies for early treatment of chikungunya patients, thereby reducing the virus-associated persistent arthritis.


Subject(s)
Antiviral Agents , Chikungunya Fever , Chikungunya virus , Zinc Acetate , Zinc Sulfate , Chikungunya virus/drug effects , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Chikungunya Fever/drug therapy , Chikungunya Fever/virology , Zinc Acetate/pharmacology , Zinc Acetate/therapeutic use , Zinc Sulfate/pharmacology , Chlorocebus aethiops , Vero Cells , Virus Internalization/drug effects , Mice , Zinc/pharmacology , Zinc/therapeutic use , Humans , Magnesium Sulfate/pharmacology , Magnesium/pharmacology , Virus Replication/drug effects , Inhibitory Concentration 50 , Salts/pharmacology , Cell Line
14.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892254

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic. Known as COVID-19, it has affected billions of people worldwide, claiming millions of lives and posing a continuing threat to humanity. This is considered one of the most extensive pandemics ever recorded in human history, causing significant losses to both life and economies globally. However, the available evidence is currently insufficient to establish the effectiveness and safety of antiviral drugs or vaccines. The entry of the virus into host cells involves binding to angiotensin-converting enzyme 2 (ACE2), a cell surface receptor, via its spike protein. Meanwhile, transmembrane protease serine 2 (TMPRSS2), a host surface protease, cleaves and activates the virus's S protein, thus promoting viral infection. Plant protease inhibitors play a crucial role in protecting plants against insects and/or microorganisms. The major storage proteins in sweet potato roots include sweet potato trypsin inhibitor (SWTI), which accounts for approximately 60% of the total water-soluble protein and has been found to possess a variety of health-promoting properties, including antioxidant, anti-inflammatory, ACE-inhibitory, and anticancer functions. Our study found that SWTI caused a significant reduction in the expression of the ACE2 and TMPRSS2 proteins, without any adverse effects on cells. Therefore, our findings suggest that the ACE2 and TMPRSS2 axis can be targeted via SWTI to potentially inhibit SARS-CoV-2 infection.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antiviral Agents , Ipomoea batatas , SARS-CoV-2 , Serine Endopeptidases , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Animals , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Ipomoea batatas/virology , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , COVID-19/virology , COVID-19/metabolism , Trypsin Inhibitors/pharmacology , Trypsin Inhibitors/metabolism , Virus Internalization/drug effects , Chlorocebus aethiops , Vero Cells , Down-Regulation/drug effects , Mice
15.
Proc Natl Acad Sci U S A ; 121(24): e2403389121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833471

ABSTRACT

Cell-cell fusion mediated by most paramyxovirus requires fusion protein (F) and attachment protein (H, HN, or G). The F protein is proteolytic cleaved to be fusogenically active. J paramyxovirus (JPV) has a unique feature in the family Paramyxoviridae: It encodes an integral membrane protein, syncytial protein (SP, formerly known as transmembrane protein, TM), which is essential in JPV-promoted cell-cell fusion (i.e., syncytial). In this study, we report that cleavage of SP is essential for its syncytial-promoting activity. We have identified the cleavage site of SP at amino acid residues 172 to 175, LKTG, and deletion of the "LKTG" residues abolished SP protein cleavage and its ability to promote cell-cell fusion. Replacing the cleavage site LKTG with a factor Xa protease cleavage site allows cleavage of the SP with factor Xa protease and restores its ability to promote cell-cell fusion. Furthermore, results from a hemifusion assay indicate that cleavage of SP plays an important role in the progression from the intermediate hemifusion state to a complete fusion. This work indicates that SP has many characteristics of a fusion protein. We propose that SP is likely a cell-cell fusion-promoting protein.


Subject(s)
Cell Fusion , Viral Fusion Proteins , Animals , Viral Fusion Proteins/metabolism , Chlorocebus aethiops , Proteolysis , Vero Cells , Virus Internalization , Factor Xa/metabolism , Humans , Cell Line
16.
PLoS Pathog ; 20(6): e1012267, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38857290

ABSTRACT

HSV infects keratinocytes in the epidermis of skin via nectin-1. We established a human foreskin explant infection model to investigate HSV entry and spread. HSV1 entry could only be achieved by the topical application of virus via high density microarray projections (HD-MAPs) to the epidermis, which penetrated beyond one third of its thickness, simulating in vivo microtrauma. Rapid lateral spread of HSV1 to a mean of 13 keratinocytes wide occurred after 24 hours and free virus particles were observed between keratinocytes, consistent with an intercellular route of spread. Nectin-1 staining was markedly decreased in foci of infection in the epidermis and in the human keratinocyte HaCaT cell line. Nectin-1 was redistributed, at the protein level, in adjacent uninfected cells surrounding infection, inducible by CCL3, IL-8 (or CXCL8), and possibly CXCL10 and IL-6, thus facilitating spread. These findings provide the first insights into HSV1 entry and spread in human inner foreskin in situ.


Subject(s)
Chemokines , Foreskin , Herpes Simplex , Herpesvirus 1, Human , Keratinocytes , Nectins , Humans , Male , Keratinocytes/virology , Keratinocytes/metabolism , Foreskin/virology , Foreskin/cytology , Nectins/metabolism , Herpes Simplex/virology , Herpes Simplex/metabolism , Chemokines/metabolism , Herpesvirus 1, Human/physiology , Cell Adhesion Molecules/metabolism , Virus Internalization
17.
Nat Commun ; 15(1): 5179, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898037

ABSTRACT

Viral genetic diversity presents significant challenges in developing antivirals with broad-spectrum activity and high barriers to resistance. Here we report development of proteolysis targeting chimeras (PROTACs) targeting the dengue virus envelope (E) protein through coupling of known E fusion inhibitors to ligands of the CRL4CRBN E3 ubiquitin ligase. The resulting small molecules block viral entry through inhibition of E-mediated membrane fusion and interfere with viral particle production by depleting intracellular E in infected Huh 7.5 cells. This activity is retained in the presence of point mutations previously shown to confer partial resistance to the parental inhibitors due to decreased inhibitor-binding. The E PROTACs also exhibit broadened spectrum of activity compared to the parental E inhibitors against a panel of mosquito-borne flaviviruses. These findings encourage further exploration of targeted protein degradation as a differentiated and potentially advantageous modality for development of broad-spectrum direct-acting antivirals.


Subject(s)
Antiviral Agents , Dengue Virus , Flavivirus , Proteolysis , Virus Internalization , Humans , Proteolysis/drug effects , Animals , Antiviral Agents/pharmacology , Flavivirus/drug effects , Flavivirus/genetics , Flavivirus/metabolism , Virus Internalization/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Dengue Virus/genetics , Culicidae/virology , Ubiquitin-Protein Ligases/metabolism , Viral Envelope Proteins/metabolism , Cell Line
18.
Cell Host Microbe ; 32(6): 945-946, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870904

ABSTRACT

In this issue of Cell Host & Microbe, Shang et al. identify murine neuropilin 1 as a host factor that binds reovirus particles, directing cell entry and contributing to viral dissemination and neurovirulence. This study highlights the reovirus model system to investigate host receptors and their significance in viral pathogenesis.


Subject(s)
Neurons , Neuropilin-1 , Reoviridae , Virus Internalization , Animals , Mice , Neurons/virology , Neuropilin-1/metabolism , Reoviridae/physiology , Reoviridae/genetics , Reoviridae/pathogenicity , Humans , Host-Pathogen Interactions , Reoviridae Infections/virology , Receptors, Virus/metabolism
19.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851803

ABSTRACT

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Subject(s)
Alphavirus , Antiviral Agents , Receptors, LDL , Virus Internalization , Animals , Humans , Alphavirus/drug effects , Alphavirus/physiology , Alphavirus/genetics , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Protein Binding , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Virion/metabolism , Virus Internalization/drug effects
20.
J Med Virol ; 96(6): e29730, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38860570

ABSTRACT

Hantaan virus (HTNV) infection can cause hemorrhagic fever with renal syndrome (HFRS) in humans, and currently, there are no long-standing protective vaccines or specific antivirals available. Guanylate-binding protein 1 (GBP1) is an interferon-stimulated gene that defends against various pathogen infections. However, the function of GBP1 in HTNV infection remains unknown. Here, we describe how GBP1 prevents HTNV infection by obstructing virus entry. We found that HTNV infection induced GBP1 expression and that overexpression of GBP1 inhibited HTNV infection, while knockout of GBP1 had the opposite effect. Interestingly, GBP1 did not affect interferon (IFN) signaling during HTNV infection. Instead, GBP1 prevented HTNV from entering cells through clathrin-mediated endocytosis (CME). We also discovered that GBP1 specifically interacted with actin but not dynamin 2 (DNM2) and made it difficult for DNM2 to be recruited by actin, which may account for the suppression of CME during HTNV infection. These findings establish an antiviral role for GBP1 in inhibiting HTNV infection and help us better understand how GBP1 regulates HTNV entry and could potentially aid in developing treatments for this virus.


Subject(s)
Endocytosis , GTP-Binding Proteins , Hantaan virus , Virus Internalization , Hantaan virus/physiology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Actins/metabolism , Host-Pathogen Interactions , Hemorrhagic Fever with Renal Syndrome/virology , Animals , Dynamin II/metabolism , Dynamin II/genetics , HEK293 Cells , Cell Line
SELECTION OF CITATIONS
SEARCH DETAIL
...