Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Int J Nanomedicine ; 19: 9009-9033, 2024.
Article in English | MEDLINE | ID: mdl-39246425

ABSTRACT

Background: The high infectivity of coronaviruses has led to increased interest in developing new strategies to prevent virus spread. Silver nanoparticles (AgNPs) and graphene oxide (GO) have attracted much attention in the antiviral field. We investigated the potential antiviral activity of GO and AgNPs combined in the nanocomposite GO-Ag against murine betacoronavirus MHV using an in vitro model. Methods: GO, AgNPs, and GO-Ag characterization (size distribution, zeta potential, TEM visualization, FT-IR, and EDX analysis) and XTT assay were performed. The antiviral activity of GO-Ag nanocomposites was evaluated by RT-qPCR and TCID50 assays. The results were compared with free AgNPs and pure GO. Cell growth and morphology of MHV-infected hepatocytes treated with GO-Ag composites were analyzed by JuLI™Br. Immunofluorescence was used to visualize the cell receptor used by MHV. Ultrastructural SEM analysis was performed to examine cell morphology after MHV infection and GO-Ag composite treatment. Results: A significant reduction in virus titer was observed for all nanocomposites tested, ranging from 3.2 to 7.3 log10 TCID50. The highest titer reduction was obtained for GO 5 µg/mL - Ag 25 µg/mL in the post-treatment method. These results were confirmed by RT-qPCR analysis. The results indicate that GO-Ag nanocomposites exhibited better antiviral activity compared to AgNPs and GO. Moreover, the attachment of AgNPs to the GO flake platform reduced their cytotoxicity. In addition, the GO-Ag composite modulates the distribution of the Ceacam1 cell receptor and can modulate cell morphology. Conclusion: Graphene oxide sheets act as a stabilizing agent, inhibiting the accumulation of AgNPs and reducing their cellular toxicity. The GO-Ag composite can physically bind and inhibit murine betacoronavirus from entering cells. Furthermore, the constant presence of GO-Ag can inhibit MHV replication and significantly limit its extracellular release. In conclusion, GO-Ag shows promise as an antiviral coating on solid surfaces to minimize virus transmission and spread.


Subject(s)
Antiviral Agents , Graphite , Metal Nanoparticles , Nanocomposites , Silver , Graphite/pharmacology , Graphite/chemistry , Silver/chemistry , Silver/pharmacology , Animals , Nanocomposites/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Mice , Metal Nanoparticles/chemistry , Murine hepatitis virus/drug effects , Hepatocytes/drug effects , Hepatocytes/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Cell Line
2.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062750

ABSTRACT

Plant polyphenols possess diverse bioactivities, including antiviral activity against a broad spectrum of viruses. Here, we investigated the virucidal properties of an Kalanchoe daigremontiana extract using an in vitro model of human herpesvirus type 1 (HHV-1) infection. Chromatographic analysis indicated that the extract of Kalanchoe daigremontiana is rich in various compounds, among which are polyphenols with virucidal activity confirmed in the literature. We found that Kalanchoe daigremontiana extract shows an ability to prevent HHV-1 infection by direct inhibition of the virus attachment, penetration, and blocking of infection when used in pretreatment or post-entry treatment. Our results indicate that Kalanchoe daigremontiana extract may be a good candidate drug against HHV-1, both as a substance to prevent infection and to treat an already ongoing infection. Our findings illustrate that Kalanchoe daigremontiana could be a potential new candidate for clinical consideration in the treatment of HHV-1 infection alone or in combination with other therapeutics.


Subject(s)
Antiviral Agents , Herpesvirus 1, Human , Kalanchoe , Plant Extracts , Plant Extracts/pharmacology , Plant Extracts/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Kalanchoe/chemistry , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Polyphenols/pharmacology , Polyphenols/chemistry , Virus Internalization/drug effects , Virus Attachment/drug effects
3.
Pathogens ; 13(2)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38392896

ABSTRACT

Human adenovirus (HAdV) is a common pathogen, which can lead to various clinical symptoms and-in some cases-central nervous system (CNS) dysfunctions, such as encephalitis and meningitis. Although the initial events of virus entry have already been identified in various cell types, the mechanism of neuronal uptake of adenoviruses is relatively little understood. The aim of this study was to investigate early events during adenoviral infection, in particular to determine the connection between cellular coxsackievirus and adenovirus receptor (CAR), clathrin, caveolin, and early endosomal proteins (EEA1 and Rab5) with the entry of HAdVs into primary murine neurons in vitro. An immunofluorescence assay and confocal microscopy analysis were carried out to determine HAdV4, 5, and 7 correlation with CAR, clathrin, caveolin, and early endosomal proteins in neurons. The quantification of Pearson's coefficient between CAR and HAdVs indicated that the HAdV4 and HAdV5 types correlated with CAR and that the correlation was more substantial for HAdV5. Inhibition of clathrin-mediated endocytosis using chlorpromazine limited the infection with HAdV, whereas inhibition of caveolin-mediated endocytosis did not affect virus entry. Thus, the entry of tested HAdV types into neurons was most likely associated with clathrin but not caveolin. It was also demonstrated that HAdVs correlate with the Rab proteins (EEA1, Rab5) present in early vesicles, and the observed differences in the manner of correlation depended on the serotype of the virus. With our research, we strove to expand knowledge regarding the mechanism of HAdV entry into neurons, which may be beneficial for developing potential therapeutics in the future.

4.
BMC Vet Res ; 20(1): 18, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195523

ABSTRACT

Nowadays, the population is still struggling with a post-COVID19 syndrome known as long COVID, including a broad spectrum of neurological problems. There is an urgent need for a better understanding and exploration of the mechanisms of coronavirus neurotropism. For this purpose, the neurotropic strain of mouse hepatitis virus (MHV-JHM) originating from the beta-coronavirus genus, the same as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been used. The role of the cytoskeleton during virus replication in neurons in vitro was determined to understand the mechanisms of MHV-JHM neuroinfection. We have described for the first time the changes of actin filaments during MHV-JHM infection. We also observed productive replication of MHV-JHM in neurons during 168 h p.i. and syncytial cytopathic effect. We discovered that the MHV-JHM strain modulated neuronal cytoskeleton during infection, which were manifested by: (i) condensation of actin filaments in the cortical layer of the cytoplasm, (ii) formation of microtubule cisternae structures containing viral antigen targeting viral replication site (iii) formation of tunneling nanotubes used by MHV-JHM for intercellular transport. Additionally, we demonstrated that the use of cytoskeletal inhibitors have reduced virus replication in neurons, especially noscapine and nocodazole, the microtubule shortening factors.


Subject(s)
COVID-19 , Murine hepatitis virus , Rodent Diseases , Animals , Mice , Post-Acute COVID-19 Syndrome/veterinary , COVID-19/veterinary , Antigens, Viral , Neurons , SARS-CoV-2
5.
Pathogens ; 11(8)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-36014997

ABSTRACT

Mitochondria are key cellular organelles responsible for many essential functions, including ATP production, ion homeostasis and apoptosis induction. Recent studies indicate their significant role during viral infection. In the present study, we examined the effects of equine herpesvirus type 1 (EHV-1) infection on the morphology and mitochondrial function in primary murine neurons in vitro. We used three EHV-1 strains: two non-neuropathogenic (Jan-E and Rac-H) and one neuropathogenic (EHV-1 26). The organization of the mitochondrial network during EHV-1 infection was assessed by immunofluorescence. To access mitochondrial function, we analyzed reactive oxygen species (ROS) production, mitophagy, mitochondrial inner-membrane potential, mitochondrial mass, and mitochondrial genes' expression. Changes in mitochondria morphology during infection suggested importance of their perinuclear localization for EHV-1 replication. Despite these changes, mitochondrial functions were preserved. For all tested EHV-1 strains, the similarities in the increased fold expression were detected only for COX18, Sod2, and Tspo. For non-neuropathogenic strains (Jan-E and Rac-H), we detected mainly changes in the expression of genes related to mitochondrial morphology and transport. The results indicate that mitochondria play an important role during EHV-1 replication in cultured neurons and undergo specific morphological and functional modifications.

6.
Pathogens ; 11(4)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35456075

ABSTRACT

Equid alphaherpesvirus 1 (EHV-1) causes respiratory diseases, abortion, and neurological disorders in horses. Recently, the oncolytic potential of this virus and its possible use in anticancer therapy has been reported, but its influence on cytoskeleton was not evaluated yet. In the following study, we have examined disruptions in actin cytoskeleton of glioblastoma multiforme in vitro model-A172 cell line, caused by EHV-1 infection. We used three EHV-1 strains: two non-neuropathogenic (Jan-E and Rac-H) and one neuropathogenic (EHV-1 26). Immunofluorescent labelling, confocal microscopy, real-time cell growth analysis and OrisTM cell migration assay revealed disturbed migration of A172 cells infected with the EHV-1, probably due to rearrangement of actin cytoskeleton and the absence of cell projections. All tested strains caused disruption of the actin network and general depolymerization of microfilaments. The qPCR results confirmed the effective replication of EHV-1. Thus, we have demonstrated, for the first time, that EHV-1 infection leads to inhibition of proliferation and migration in A172 cells, which might be promising for new immunotherapy treatment.

7.
Viruses ; 13(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34696425

ABSTRACT

Sialodacryoadenitis virus (SDAV) is known to be an etiological agent, causing infections in laboratory rats. Until now, its role has only been considered in studies on respiratory and salivary gland infections. The scant literature data, consisting mainly of papers from the last century, do not sufficiently address the topic of SDAV infections. The ongoing pandemic has demonstrated, once again, the role of the Coronaviridae family as extremely dangerous etiological agents of human zoonoses. The ability of coronaviruses to cross the species barrier and change to hosts commonly found in close proximity to humans highlights the need to characterize SDAV infections. The main host of the infection is the rat, as mentioned above. Rats inhabit large urban agglomerations, carrying a vast epidemic threat. Of the 2277 existing rodent species, 217 are reservoirs for 66 zoonotic diseases caused by viruses, bacteria, fungi, and protozoa. This review provides insight into the current state of knowledge of SDAV characteristics and its likely zoonotic potential.


Subject(s)
Coronavirus Infections/veterinary , Coronavirus, Rat/genetics , Coronavirus, Rat/pathogenicity , Viral Zoonoses/epidemiology , Animals , Coronavirus Infections/transmission , Coronavirus, Rat/classification , Rats , Species Specificity , Virus Replication/physiology
8.
Arch Virol ; 166(5): 1371-1383, 2021 May.
Article in English | MEDLINE | ID: mdl-33715038

ABSTRACT

Herpesviruses are capable of infecting not only neurons, where they establish latent infection, but also astrocytes. Since astrocytes are important for the functioning of the central nervous system (CNS), their infection may lead to serious neurological disorders. Thus, in the present study we investigated the ability of human herpesvirus type 2 (HHV-2) to infect primary murine astrocytes in vitro and the effect of infection on their mitochondrial network and actin cytoskeleton. In immunofluorescence assays, antibodies against HHV-2 antigens and glial fibrillary acidic protein (GFAP) were used to confirm that the infected cells are indeed astrocytes. Real-time PCR analysis showed a high level of HHV-2 replication in astrocytes, particularly at 168 h postinfection, confirming that a productive infection had occurred. Analysis of mitochondrial morphology showed that, starting from the first stage of infection, HHV-2 caused fragmentation of the mitochondrial network and formation of punctate and tubular structures that colocalized with virus particles. Furthermore, during the late stages of infection, the infection affected the actin cytoskeleton and induced formation of actin-based cellular projections, which were probably associated with enhanced intracellular spread of the virus. These results suggest that the observed changes in the mitochondrial network and actin cytoskeleton in productively infected astrocytes are required for effective replication and viral spread in a primary culture of astrocytes. Moreover, we speculate that, in response to injury such as HHV-2 infection, murine astrocytes cultured in vitro undergo transformation, defined in vivo as reactive astrocytosis.


Subject(s)
Actin Cytoskeleton/pathology , Astrocytes/virology , Herpesvirus 2, Human/physiology , Mitochondria/pathology , Actin Cytoskeleton/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Gliosis , Kinetics , Mice , Mitochondria/metabolism , Virion/metabolism , Virus Replication
9.
J Neurovirol ; 25(6): 765-782, 2019 12.
Article in English | MEDLINE | ID: mdl-31161588

ABSTRACT

Human herpesvirus types 1 and 2 (HHV-1 and HHV-2) are neurotropic viruses which remain latent for life and reactivate to cause recurrent infections. HHV-1 has been found to be involved in accumulation of ß-amyloid, hyperphosphorylation of tau proteins, and inflammation in the brain, which can later result in neuronal dysfunction and neurodegeneration. The relationship between HHV-2 and events associated with neurodegeneration has not been extensively studied. Neurons, more than any other cell type, depend on mitochondrial trafficking for their survival, and many types of mitochondrial abnormalities have been described in the etiology of neurodegenerative diseases. Therefore, in this study, we concentrated on mitochondrial dysfunction associated with HHV-1 and HHV-2 infection of primary murine neurons in vitro. We showed that starting from the first stages of HHV-1 and HHV-2 infection, an interaction of viral particles with the mitochondrial network occurs. Both HHV-1 and HHV-2 infection affected mitochondrial function at multiple levels, including upregulation of mitochondrial fission, decrease of the mitochondrial membrane potential, and increase of ROS level. The changes observed in the organization of the mitochondrial network and physiology of productively infected neurons provide appropriate conditions for HHV-1 and HHV-2 replication and are required for effective viral spread.


Subject(s)
Herpesviridae Infections/virology , Mitochondria/virology , Mitochondrial Dynamics , Neurons/metabolism , Neurons/virology , Animals , Cells, Cultured , Herpesviridae Infections/metabolism , Herpesvirus 1, Human , Herpesvirus 2, Human , Mice , Mice, Inbred BALB C , Mitochondria/metabolism
10.
Arch Virol ; 163(10): 2663-2673, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29872950

ABSTRACT

Mitochondrial movement and distribution throughout the cytoplasm is crucial for maintaining cell homeostasis. Mitochondria are dynamic organelles but can be functionally disrupted during infection. Here, we show that the ubiquitous human pathogens HHV-1 and HHV-2 induce changes in the mitochondrial morphology and distribution in the early and late phases of productive infection in human keratinocytes (HaCaT cells). We observed a decrease in the mitochondrial potential at 2 h postinfection and a decrease in cell vitality at 24 h postinfection. Moreover, we found that mitochondria migrated to the perinuclear area, where HHV-1 and HHV-2 antigens were also observed, mainly in the early stages of infection. Positive results of real-time PCR showed a high level of HHV-1 and HHV-2 DNA in HaCaT cells and culture medium. Our data demonstrate that HHV-1 and HHV-2 cause mitochondrial dysfunction in human keratinocytes.


Subject(s)
Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Keratinocytes/pathology , Mitochondria/pathology , Mitochondrial Dynamics/physiology , Antigens, Viral/immunology , Cell Line, Transformed , Cell Movement , DNA, Viral/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Humans , Mitochondria/virology
11.
Postepy Hig Med Dosw (Online) ; 70(0): 572-80, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-27333927

ABSTRACT

Viruses, despite being relatively simple in structure and composition, have evolved a broad spectrum of mechanisms to exploit the host cell. To initiate effective infection, viruses or viral genomes have to enter cells. Recently studies have shown that apart from the direct fusion at the plasma membrane, endocytosis is more often the preferred means of entry into the host cell. Endocytosis is a complex phenomenon, that includes multiple pathways of membrane trafficking, such as clathrin-mediated endocytosis, caveolin-mediated endocytosis, macropinocytosis and phagocytosis. Endosomes offer a convenient and often rapid transit system across the plasma membrane and cytoplasm via the cellular microtubular network. They also provide protection to the virus from detection by the host's innate immune defences. What is important, viruses are able to utilize not just one, but multiple uptake routes. Identification of these processes and factors will not only allow a better insight into pathogenic mechanism, but may identify novel targets for future therapeutic development. This review provides insight on recent developments in the rapidly evolving field of viral entry.


Subject(s)
Endocytosis , Host-Pathogen Interactions , Virus Diseases/physiopathology , Virus Internalization , Humans
SELECTION OF CITATIONS
SEARCH DETAIL