Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Ther Oncolytics ; 31: 100743, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38033400

RESUMO

Wild-type reovirus serotype 3 Dearing (T3wt), a non-pathogenic intestinal virus, has shown promise as a cancer therapy in clinical trials, but it would benefit from an increased potency. Given that T3wt is naturally adapted to the intestinal environment (rather than tumors), we genetically modified reovirus to improve its infectivity in cancer cells. Various reovirus mutants were created, and their oncolytic potency was evaluated in vitro using plaque size as a measure of virus fitness in cancer cells. Notably, Super Virus 5 (SV5), carrying five oncolytic mutations, displayed the largest plaques in breast cancer cells among the mutants tested, indicating the potential for enhancing oncolytic potency through the combination of mutations. Furthermore, in a HER2+ murine breast cancer model, mice treated with SV5 exhibited superior tumor reduction and increased survival compared with those treated with PBS or T3wt. Intriguingly, SV5 did not replicate faster than T3wt in cultured cells but demonstrated a farther spread relative to T3wt, attributed to its reduced attachment to cancer cells. These findings highlight the significance of increased virus spread as a crucial mechanism for improving oncolytic virus activity. Thus, genetic modifications of reovirus hold the potential for augmenting its efficacy in cancer therapy.

2.
Adv Colloid Interface Sci ; 320: 103006, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37778249

RESUMO

While SARS-CoV-2 is generally under control, the question of variants and infections still persists. Fundamental information on how the virus interacts with inanimate surfaces commonly found in our daily life and when in contact with the skin will be helpful in developing strategies to inhibit the spread of the virus. Here in, a critically important review of current understanding of the interaction between virus and surface is summarized from chemistry point-of-view. The Derjaguin-Landau-Verwey-Overbeek and extended Derjaguin-Landau-Verwey-Overbeek theories to model virus attachments on surfaces are introduced, along with the interaction type and strength, and quantification of each component. The virus survival and transfer are affected by a combination of biological, physical, and chemical parameters, as well as environmental parameters. The surface properties for virus and virus survival on typical surfaces such as metals, plastics, and glass are summarized. Attention is also paid to the transfer of virus to/from surfaces and skin. Typical virus disinfection strategies utilizing heat, light, chemicals, and ozone are discussed together with their disinfection mechanism. In the last section, design principles for virus repelling surface chemistry such as surperhydrophobic or surperhydrophilic surfaces are also introduced, to demonstrate how the integration of surface property control and advanced material fabrication can lead to the development of functional surfaces for mitigating the effect of viral infection upon contact.


Assuntos
Desinfecção , Propriedades de Superfície
3.
Pharmaceuticals (Basel) ; 16(2)2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-37259408

RESUMO

BACKGROUND: At present, about half of the world's population is at risk of being infected with dengue virus (DENV). However, there are no specific drugs to prevent or treat DENV infection. Glycyrrhizae Radix et Rhizome, a well-known traditional Chinese medicine, performs multiple pharmacological activities, including exerting antiviral effects. The aim of this study was to investigate the anti-DENV effects of n-butanol extract from Glycyrrhizae Radix et Rhizome (GRE). METHODS: Compounds analysis of GRE was conducted via ultra-performance liquid chromatography/tandem mass spectrometry (UHPLC-MS/MS). The antiviral activities of GRE were determined by the CCK-8 assay, plaque assay, qRT-PCR, Western blotting, and the immunofluorescence assay. The DENV-infected suckling mice model was constructed to explore the antiviral effects of GRE in vivo. RESULTS: Four components in GRE were analyzed by UHPLC-MS/MS, including glycyrrhizic acid, glycyrrhetnic acid, liquiritigenin, and isoliquiritigenin. GRE inhibited the attachment process of the virus replication cycle and reduced the expression of the E protein in cell models. In the in vivo study, GRE significantly relieved clinical symptoms and prolong survival duration. GRE also significantly decreased viremia, reduced the viral load in multiple organs, and inhibited the release of pro-inflammatory cytokines in DENV-infected suckling mice. CONCLUSIONS: GRE exhibited significant inhibitory activities in the adsorption stage of the DENV-2 replication cycle by targeting the envelope protein. Thus, GRE might be a promising candidate for the treatment of DENV infection.

4.
Front Vet Sci ; 9: 978453, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36061121

RESUMO

Swine acute diarrhea syndrome coronavirus (SADS-CoV) is an emerging swine enteropathogenic coronavirus that causes severe diarrhea in neonatal piglets, leading to serious economic losses to the pig industries. At present, there are no effective control measures for SADS, making an urgent need to exploit effective antiviral therapies. Here, we confirmed that Aloe extract (Ae) can strongly inhibit SADS-CoV in Vero and IPI-FX cells in vitro. Furthermore, we detected that Emodin from Ae had anti-SADS-CoV activity in cells but did not impair SADS-CoV infectivity directly. The time-of-addition assay showed that Emodin inhibits SADS-CoV infection at the whole stages of the viral replication cycle. Notably, we found that Emodin can significantly reduce virus particles attaching to the cell surface and induce TLR3 (p < 0.001), IFN-λ3 (p < 0.01), and ISG15 (p < 0.01) expressions in IPI-FX cells, indicating that the anti-SADS-CoV activity of Emodin might be due to blocking viral attachment and the activation of TLR3-IFN-λ3-ISG15 signaling axis. These results suggest that Emodin has the potential value for the development of anti-SADS-CoV drugs.

5.
Water Res ; 219: 118596, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35598470

RESUMO

Effective removal of human pathogenic viruses is an indispensable yet rarely studied aspect for sustainable treatment of domestic wastewater by anaerobic membrane bioreactor (AnMBR). In this study, the interface behaviors and removal mechanisms of norovirus genogroup I (GI), genogroup II (GII), and rotavirus A from domestic wastewater was systematically investigated in a one-stage AnMBR. On average, norovirus GI, GII and rotavirus were reduced by 4.64, 5.00, and 2.31 logs, respectively. Viruses tended to be transferred to larger-sized suspended solids from sewage influent to the mixed liquor, and the weight-specific concentration of the virus in >100 µm particles of the mixed liquor was significantly higher than that of sewage, indicating a particle scale-dependent affinity with the virus. In-series membrane filtration test showed the main contribution of the membrane retention, which was dominated by the bio-cake layer and the pristine membrane, while the membrane and associated pore foulants can retain viruses in a filtration resistance-efficient way. An unsteady-state mass balance model revealed that free viruses in the bulk liquid of AnMBR were minimally attached to the cake layer but mainly retained by the membrane and pore foulants (>99%). In addition, despite the small virus decay rates in the mixed liquor, the associated contribution increased with run time due to the prolonged sludge retention time. These insights into virus behaviors and removal mechanisms may provide novel regulation strategies for enhanced virus removal by AnMBR.


Assuntos
Norovirus , Vírus , Anaerobiose , Reatores Biológicos , Humanos , Membranas Artificiais , Esgotos , Eliminação de Resíduos Líquidos/métodos , Águas Residuárias
6.
Viruses ; 13(11)2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34835112

RESUMO

Anti-viral small molecules are currently lacking for treating coronavirus infection. The long development timescales for such drugs are a major problem, but could be shortened by repurposing existing drugs. We therefore screened a small library of FDA-approved compounds for potential severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antivirals using a pseudovirus system that allows a sensitive read-out of infectivity. A group of structurally-related compounds, showing moderate inhibitory activity with IC50 values in the 2-5 µM range, were identified. Further studies demonstrated that these "kite-shaped" molecules were surprisingly specific for SARS-CoV-1 and SARS-CoV-2 and that they acted early in the entry steps of the viral infectious cycle, but did not affect virus attachment to the cells. Moreover, the compounds were able to prevent infection in both kidney- and lung-derived human cell lines. The structural homology of the hits allowed the production of a well-defined pharmacophore that was found to be highly accurate in predicting the anti-viral activity of the compounds in the screen. We discuss the prospects of repurposing these existing drugs for treating current and future coronavirus outbreaks.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , COVID-19/virologia , Vírus da Leucemia Murina/efeitos dos fármacos , SARS-CoV-2 , Internalização do Vírus/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos , Sinergismo Farmacológico , Humanos , Vírus da Leucemia Murina/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Ligação Viral/efeitos dos fármacos
7.
Viruses ; 13(10)2021 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-34696365

RESUMO

Nascent HIV-1 particles incorporate the viral envelope glycoprotein and multiple host transmembrane proteins during assembly at the plasma membrane. At least some of these host transmembrane proteins on the surface of virions are reported as pro-viral factors that enhance virus attachment to target cells or facilitate trans-infection of CD4+ T cells via interactions with non-T cells. In addition to the pro-viral factors, anti-viral transmembrane proteins are incorporated into progeny virions. These virion-incorporated transmembrane proteins inhibit HIV-1 entry at the point of attachment and fusion. In infected polarized CD4+ T cells, HIV-1 Gag localizes to a rear-end protrusion known as the uropod. Regardless of cell polarization, Gag colocalizes with and promotes the virion incorporation of a subset of uropod-directed host transmembrane proteins, including CD162, CD43, and CD44. Until recently, the functions of these virion-incorporated proteins had not been clear. Here, we review the recent findings about the roles played by virion-incorporated CD162, CD43, and CD44 in HIV-1 spread to CD4+ T cells.


Assuntos
Infecções por HIV/metabolismo , Receptores de Hialuronatos/metabolismo , Leucossialina/metabolismo , Glicoproteínas de Membrana/metabolismo , Membrana Celular/metabolismo , Infecções por HIV/genética , HIV-1/genética , HIV-1/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Receptores de Hialuronatos/genética , Leucossialina/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Linfócitos T/metabolismo , Linfócitos T/virologia , Vírion/metabolismo , Montagem de Vírus , Ligação Viral , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
8.
Nanomaterials (Basel) ; 11(1)2021 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-33435619

RESUMO

Controlled deposition of nanoparticles and bioparticles is necessary for their separation and purification by chromatography, filtration, food emulsion and foam stabilization, etc. Compared to numerous experimental techniques used to quantify bioparticle deposition kinetics, the quartz crystal microbalance (QCM) method is advantageous because it enables real time measurements under different transport conditions with high precision. Because of its versatility and the deceptive simplicity of measurements, this technique is used in a plethora of investigations involving nanoparticles, macroions, proteins, viruses, bacteria and cells. However, in contrast to the robustness of the measurements, theoretical interpretations of QCM measurements for a particle-like load is complicated because the primary signals (the oscillation frequency and the band width shifts) depend on the force exerted on the sensor rather than on the particle mass. Therefore, it is postulated that a proper interpretation of the QCM data requires a reliable theoretical framework furnishing reference results for well-defined systems. Providing such results is a primary motivation of this work where the kinetics of particle deposition under diffusion and flow conditions is discussed. Expressions for calculating the deposition rates and the maximum coverage are presented. Theoretical results describing the QCM response to a heterogeneous load are discussed, which enables a quantitative interpretation of experimental data obtained for nanoparticles and bioparticles comprising viruses and protein molecules.

9.
Artigo em Inglês | MEDLINE | ID: mdl-33495228

RESUMO

Herpesviruses are widespread and can cause serious illness. Many currently available antiviral drugs have limited effects, result in rapid development of resistance, and often exhibit dose-dependent toxicity. Especially for human cytomegalovirus (HCMV), new well-tolerated compounds with novel mechanisms of action are urgently needed. In this study, we characterized the antiviral activity of two new diazadispiroalkane derivatives, 11826091 and 11826236. These two small molecules exhibited strong activity against low-passage-number HCMV. Pretreatment of cell-free virus with these compounds greatly reduced infection. Time-of-addition assays where 11826091 or 11826236 was added to cells before infection, before and during infection, or during or after infection demonstrated an inhibitory effect on early steps of infection. Interestingly, 11826236 had an effect by addition to cells after infection. Results from entry assays showed the major effect to be on attachment. Only 11826236 had a minimal effect on penetration comparable to heparin. Further, no effect on virus infection was found for cell lines with a defect in heparan sulfate expression or lacking all surface glycosaminoglycans, indicating that these small molecules bind to heparan sulfate on the cell surface. To test this further, we extended our analyses to pseudorabies virus (PrV), a member of the Alphaherpesvirinae, which is known to use cell surface heparan sulfate for initial attachment via nonessential glycoprotein C (gC). While infection with PrV wild type was strongly impaired by 11826091 or 11826236, as with heparin, a mutant lacking gC was unaffected by either treatment, demonstrating that primary attachment to heparan sulfate via gC is targeted by these small molecules.


Assuntos
Herpesvirus Suídeo 1 , Internalização do Vírus , Alcanos , Animais , Antivirais , Glicosaminoglicanos , Heparina/farmacologia , Heparitina Sulfato , Humanos , Compostos de Espiro , Proteínas do Envelope Viral
10.
J Virol ; 95(6)2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33361418

RESUMO

Dabbling and diving ducks partly occupy shared habitats but have been reported to play different roles in wildlife infectious disease dynamics. Influenza A virus (IAV) epidemiology in wild birds has been based primarily on surveillance programs focused on dabbling duck species, particularly mallard (Anas platyrhynchos). Surveillance in Eurasia has shown that in mallards, some subtypes are commonly (H1 to H7 and H10), intermediately (H8, H9, H11, and H12), or rarely (H13 to H16) detected, contributing to discussions on virus host range and reservoir competence. An alternative to surveillance in determining IAV host range is to study virus attachment as a determinant for infection. Here, we investigated the attachment patterns of all avian IAV subtypes (H1 to H16) to the respiratory and intestinal tracts of four dabbling duck species (Mareca and Anas spp.), two diving duck species (Aythya spp.), and chicken, as well as to a panel of 65 synthetic glycan structures. We found that IAV subtypes generally showed abundant attachment to colon of the Anas duck species, mallard, and Eurasian teal (Anas crecca), supporting the fecal-oral transmission route in these species. The reported glycan attachment profile did not explain the virus attachment patterns to tissues but showed significant attachment of duck-originated viruses to fucosylated glycan structures and H7 virus tropism for Neu5Gc-LN. Our results suggest that Anas ducks play an important role in the ecology and epidemiology of IAV. Further knowledge on virus tissue attachment, receptor distribution, and receptor binding specificity is necessary to understand the mechanisms underlying host range and epidemiology of IAV.IMPORTANCE Influenza A viruses (IAVs) circulate in wild birds worldwide. From wild birds, the viruses can cause outbreaks in poultry and sporadically and indirectly infect humans. A high IAV diversity has been found in mallards (Anas platyrhynchos), which are most often sampled as part of surveillance programs; meanwhile, little is known about the role of other duck species in IAV ecology and epidemiology. In this study, we investigated the attachment of all avian IAV hemagglutinin (HA) subtypes (H1 to H16) to tissues of six different duck species and chicken as an indicator of virus host range. We demonstrated that the observed virus attachment patterns partially explained reported field prevalence. This study demonstrates that dabbling ducks of the Anas genus are potential hosts for most IAV subtypes, including those infecting poultry. This knowledge is useful to target the sampling of wild birds in nature and to further study the interaction between IAVs and birds.


Assuntos
Patos/virologia , Vírus da Influenza A/fisiologia , Influenza Aviária/virologia , Animais , Galinhas/virologia , Colo/virologia , Patos/classificação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Especificidade de Hospedeiro , Vírus da Influenza A/classificação , Vírus da Influenza A/metabolismo , Influenza Aviária/transmissão , Polissacarídeos/química , Polissacarídeos/metabolismo , Sistema Respiratório/virologia , Tropismo Viral , Ligação Viral
11.
Eur J Med Chem ; 211: 113014, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33218683

RESUMO

Viruses are obligate intracellular parasites and have evolved to enter the host cell. To gain access they come into contact with the host cell through an initial adhesion, and some viruses from different genus may use heparan sulfate proteoglycans for it. The successful inhibition of this early event of the infection by synthetic molecules has always been an attractive target for medicinal chemists. Numerous reports have yielded insights into the function of compounds based on the dispirotripiperazine scaffold. Analysis suggests that this is a structural requirement for inhibiting the interactions between viruses and cell-surface heparan sulfate proteoglycans, thus preventing virus entry and replication. This review summarizes our current knowledge about the early history of development, synthesis, structure-activity relationships and antiviral evaluation of dispirotripiperazine-based compounds and where they are going in the future.


Assuntos
Antivirais/farmacologia , Desenho de Fármacos , Piperazinas/farmacologia , Compostos de Espiro/farmacologia , Vírus/efeitos dos fármacos , Antivirais/química , Proteoglicanas de Heparan Sulfato/antagonistas & inibidores , Proteoglicanas de Heparan Sulfato/metabolismo , Estrutura Molecular , Piperazinas/química , Compostos de Espiro/química , Vírus/metabolismo
12.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33115870

RESUMO

Human noroviruses are the most common nonbacterial cause of gastroenteritis outbreaks, with new variants and genotypes frequently emerging. The origin of these new viruses is unknown; however, animals have been proposed as a potential source, as human noroviruses have been detected in animal species. Here, we investigated the potential of animals to serve as a reservoir of human noroviruses by testing norovirus attachment to formalin-fixed intestinal tissues of a range of potential reservoir animals. We set up a novel method to study norovirus binding using fluorescein isothiocyanate (FITC)-labeled virus-like particles (VLPs). In humans, noroviruses interact with histo-blood group antigens (HBGAs), carbohydrates that are expressed, among others, on the epithelial lining of the gastrointestinal tract. In animals, this interaction is not well understood. To test if virus binding depends on HBGAs, we characterized the HBGA phenotype in animal tissues by immunohistochemistry. With the exception of the black-headed gull and the straw-colored fruitbat, we observed the attachment of several human norovirus genotypes to the intestinal epithelium of all tested animal species. However, we did not find an association between the expression of a specific HBGA phenotype and virus-like particle (VLP) attachment. We show that selected human noroviruses can attach to small-intestinal tissues across species, supporting the hypothesis that human noroviruses can reside in an animal reservoir. However, whether this attachment can subsequently lead to infection needs to be further assessed.IMPORTANCE Noroviruses are a major cause of acute gastroenteritis in humans. New norovirus variants and recombinants (re)emerge regularly in the human population. From animal experiments and surveillance studies, it has become clear that at least seven animal models are susceptible to infection with human strains and that domesticated and wild animals shed human noroviruses in their feces. As virus attachment is an important first step for infection, we used a novel method utilizing FITC-labeled VLPs to test for norovirus attachment to intestinal tissues of potential animal hosts. We further characterized these tissues with regard to their HBGA expression, a well-studied norovirus susceptibility factor in humans. We found attachment of several human strains to a variety of animal species independent of their HBGA phenotype. This supports the hypothesis that human strains could reside in an animal reservoir.


Assuntos
Antígenos de Grupos Sanguíneos/metabolismo , Infecções por Caliciviridae/virologia , Modelos Animais de Doenças , Gastroenterite/virologia , Mucosa Intestinal/virologia , Norovirus/fisiologia , Ligação Viral , Sequência de Aminoácidos , Animais , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/patologia , Fezes/virologia , Gastroenterite/metabolismo , Gastroenterite/patologia , Humanos , Mucosa Intestinal/metabolismo , Homologia de Sequência
13.
Fish Shellfish Immunol ; 107(Pt A): 9-15, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32976972

RESUMO

Tiger frog virus (TFV) belongs to the genus Ranavirus (family Iridoviridae) and causes significant harm in cultured frogs, resulting in substantial losses in ecological and economic field in Southern China. Attachment is the first step in viral life cycle, which is dependent on the interactions of virions with extracellular matrix (ECM) components. Studying this process will help in understanding virus infection and controlling viral diseases. In this study, the roles of primary ECM components in TFV attachment were investigated. The results on the kinetics of virus attachment showed TFV successful attachment to the cell surface as a relatively rapid process after TFV was used to inoculate cells for 10 min at 4 °C. Western blot and quantitative PCR analyses results showed that soluble fibronectin, collagen IV, laminin, or hyaluronic acid treatment with TFV caused no significant effect on virus attachment. Soluble heparin, heparan sulfate and chondroitin sulfate A/B could inhibit TFV attachment in a dose-dependent manner. Enzymic digestion by cell surface heparin/heparan sulfate using heparinase I, II, and III could significantly prevent TFV attachment, suggesting that heparan sulfate plays an important role in TFV attachment. Furthermore, the binding assays of heparin-agarose beads and virion showed that TFV virions specifically bound with heparin in a dose-dependent manner. Given that heparin is a structural analogue of heparan sulfate, the above results suggest that heparan sulfate might serve as an attachment factor of TFV infection. Our work would be beneficial to understand the mechanisms of TFV attachment and the interactions of TFV with cellular receptor(s).


Assuntos
Cyprinidae , Infecções por Vírus de DNA/veterinária , Doenças dos Peixes/virologia , Ranavirus/fisiologia , Ligação Viral , Animais , Linhagem Celular , Infecções por Vírus de DNA/virologia , Matriz Extracelular/fisiologia
15.
Proc Natl Acad Sci U S A ; 117(14): 8055-8063, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32193343

RESUMO

HIV-1 particles incorporate various host transmembrane proteins in addition to viral Env glycoprotein during assembly at the plasma membrane. In polarized T cells, HIV-1 structural protein Gag localizes to the plasma membrane of uropod, a rear-end protrusion. Notably, uropod transmembrane proteins PSGL-1 and CD43 cocluster specifically with Gag assembling at the plasma membrane even in cells that do not form uropods. Recent reports have shown that expression of either PSGL-1 or CD43 in virus-producing cells reduces the infectivity of progeny virions and that HIV-1 infection reduces the cell surface expression of these proteins. However, the mechanisms for both processes remain to be determined. In this study, we found that virion incorporation of PSGL-1 and CD43 closely correlates with diminished virion infectivity. PSGL-1 and CD43 inhibited virus attachment to CD4+ cells irrespective of the presence of Env. These proteins also inhibited virion attachment to CD4- lymphoid organ fibroblastic reticular cells that mediate transinfection of CD4+ T cells. Consistent with the possibility that highly extended extracellular domains of these proteins physically block virus-cell attachment, the inhibitory effect of PSGL-1 required its full-length ectodomain. HIV-1 encoding Gag mutants that are defective in either coclustering with these host proteins or ESCRT-dependent particle release failed to reduce PSGL-1 on surface of infected cells. This study reveals an anti-HIV-1 mechanism that suppresses virus-cell attachment and a previously unappreciated process of HIV-1-mediated down-regulation of host antiviral proteins, both of which likely require virion incorporation of these proteins.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Interações Hospedeiro-Patógeno/genética , Leucossialina/genética , Glicoproteínas de Membrana/genética , Vírion/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Buffy Coat/citologia , Regulação para Baixo , Técnicas de Inativação de Genes , Células HEK293 , Infecções por HIV/virologia , HIV-1/genética , HIV-1/metabolismo , Células HeLa , Voluntários Saudáveis , Interações Hospedeiro-Patógeno/imunologia , Humanos , Mutação , Domínios Proteicos/genética , Linfócitos T/imunologia , Montagem de Vírus/genética , Montagem de Vírus/imunologia , Ligação Viral , Replicação Viral/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
16.
Viruses ; 12(2)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32033244

RESUMO

The green tea catechin epigallocatechin gallate (EGCG) exhibits antiviral activity against various viruses. Whether EGCG also inhibits the infectivity of circovirus remains unclear. In this study, we demonstrated the antiviral effect of EGCG on porcine circovirus type 2 (PCV2). EGCG targets PCV2 virions directly and blocks the attachment of virions to host cells. The microscale thermophoresis assay showed EGCG could interact with PCV2 capsid protein in vitro with considerable affinity (Kd = 98.03 ± 4.76 µM), thereby interfering with the binding of the capsid to the cell surface receptor heparan sulfate. The molecular docking analysis of capsid-EGCG interaction identified the key amino acids which formed the binding pocket accommodating EGCG. Amino acids ARG51, ASP70, ARG73 and ASP78 of capsid were found to be critical for maintaining the binding, and the arginine residues were also essential for the electrostatic interaction with heparan sulfate. The rescued mutant viruses also confirm the importance of the key amino acids of the capsid to the antiviral effect of EGCG. Our findings suggest that catechins could act as anti-infective agents against circovirus invasion, as well as provide the basic information for the development and synthesis of structure-based anti-circovirus drugs.


Assuntos
Antivirais/farmacologia , Capsídeo/metabolismo , Catequina/análogos & derivados , Circovirus/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos , Animais , Capsídeo/química , Capsídeo/efeitos dos fármacos , Catequina/farmacologia , Linhagem Celular , Circovirus/classificação , Simulação de Acoplamento Molecular , Suínos , Chá/química
17.
Artigo em Inglês | MEDLINE | ID: mdl-31932383

RESUMO

Here, we report a class of tryptophan trimers and tetramers that inhibit (at low micromolar range) dengue and Zika virus infection in vitro These compounds (AL family) have three or four peripheral tryptophan moieties directly linked to a central scaffold through their amino groups; thus, their carboxylic acid groups are free and exposed to the periphery. Structure-activity relationship (SAR) studies demonstrated that the presence of extra phenyl rings with substituents other than COOH at the N1 or C2 position of the indole side chain is a requisite for the antiviral activity against both viruses. The molecules showed potent antiviral activity, with low cytotoxicity, when evaluated on different cell lines. Moreover, they were active against laboratory and clinical strains of all four serotypes of dengue virus as well as a selected group of Zika virus strains. Additional mechanistic studies performed with the two most potent compounds (AL439 and AL440) demonstrated an interaction with the viral envelope glycoprotein (domain III) of dengue 2 virus, preventing virus attachment to the host cell membrane. Since no antiviral agent is approved at the moment against these two flaviviruses, further pharmacokinetic studies with these molecules are needed for their development as future therapeutic/prophylactic drugs.


Assuntos
Antivirais/química , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Triptofano/química , Triptofano/farmacologia , Zika virus/efeitos dos fármacos , Animais , Chlorocebus aethiops , Vírus da Dengue/patogenicidade , Células Endoteliais da Veia Umbilical Humana , Humanos , Sorogrupo , Relação Estrutura-Atividade , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral/efeitos dos fármacos , Zika virus/patogenicidade
18.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31462562

RESUMO

Reovirus is undergoing clinical testing as an oncolytic therapy for breast cancer. Given that reovirus naturally evolved to thrive in enteric environments, we sought to better understand how breast tumor microenvironments impinge on reovirus infection. Reovirus was treated with extracellular extracts generated from polyomavirus middle T-antigen-derived mouse breast tumors. Unexpectedly, these breast tumor extracellular extracts inactivated reovirus, reducing infectivity of reovirus particles by 100-fold. Mechanistically, inactivation was attributed to proteolytic cleavage of the viral cell attachment protein σ1, which diminished virus binding to sialic acid (SA)-low tumor cells. Among various specific protease class inhibitors and metal ions, EDTA and ZnCl2 effectively modulated σ1 cleavage, indicating that breast tumor-associated zinc-dependent metalloproteases are responsible for reovirus inactivation. Moreover, media from MCF7, MB468, MD-MB-231, and HS578T breast cancer cell lines recapitulated σ1 cleavage and reovirus inactivation, suggesting that inactivation of reovirus is shared among mouse and human breast cancers and that breast cancer cells by themselves can be a source of reovirus-inactivating proteases. Binding assays and quantification of SA levels on a panel of cancer cells showed that truncated σ1 reduced virus binding to cells with low surface SA. To overcome this restriction, we generated a reovirus mutant with a mutation (T249I) in σ1 that prevents σ1 cleavage and inactivation by breast tumor-associated proteases. The mutant reovirus showed similar replication kinetics in tumorigenic cells, toxicity equivalent to that of wild-type reovirus in a severely compromised mouse model, and increased tumor titers. Overall, the data show that tumor microenvironments have the potential to reduce infectivity of reovirus.IMPORTANCE We demonstrate that metalloproteases in breast tumor microenvironments can inactivate reovirus. Our findings expose that tumor microenvironment proteases could have a negative impact on proteinaceous cancer therapies, such as reovirus, and that modification of such therapies to circumvent inactivation by tumor metalloproteases merits consideration.


Assuntos
Proteínas do Capsídeo/metabolismo , Infecções por Reoviridae/metabolismo , Replicação Viral/genética , Células A549 , Animais , Neoplasias da Mama/terapia , Neoplasias da Mama/virologia , Proteínas do Capsídeo/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Feminino , Células HeLa , Humanos , Metaloproteases/metabolismo , Camundongos , Mutação , Ácido N-Acetilneuramínico/metabolismo , Terapia Viral Oncolítica/métodos , Receptores Virais/metabolismo , Reoviridae/metabolismo , Reoviridae/patogenicidade , Infecções por Reoviridae/imunologia , Microambiente Tumoral/fisiologia , Proteínas Virais/metabolismo , Ligação Viral , Replicação Viral/fisiologia
19.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31167919

RESUMO

Hand, foot, and mouth disease (HFMD), a highly contagious disease in children, is caused by human enteroviruses, including enterovirus 71 (EV71), coxsackievirus A16 (CVA16), and coxsackievirus A6 (CVA6). Although HFMD is usually mild and self-limiting, EV71 infection occasionally leads to fatal neurological disorders. Currently, no commercial antiviral drugs for HFMD treatment are available. Here, numerous sulfonated azo dyes, widely used as food additives, were identified as having potent antiviral activities against human enteroviruses. Among them, brilliant black BN (E151) was able to inhibit all EV71, CVA16, and CVA6 strains tested. In rhabdomyosarcoma cells, the 50% inhibitory concentrations of the dye E151 for various strains of EV71 ranged from 2.39 µM to 28.12 µM, whereas its 50% cytotoxic concentration was 1,870 µM. Food azo dyes, including E151, interacted with the vertex of the 5-fold axis of EV71 and prevented viral entry. Their efficacy in viral inhibition was regulated by amino acids at VP1-98, VP1-145, and/or VP1-246. Dye E151 not only prevented EV71 attachment but also eluted attached viruses in a concentration-dependent manner. Moreover, E151 inhibited the interaction between EV71 and its cellular uncoating factor cyclophilin A. In vivo studies demonstrated that E151 at a dose of 200 mg/kg of body weight/day given on the initial 4 days of challenge protected AG129 mice challenged with 10× the 50% lethal dose of wild-type EV71 isolates. Taken together, these data highlight E151 as a promising antiviral agent against EV71 infection.IMPORTANCE Human enterovirus 71 (EV71) is one of the causative agents of hand, foot, and mouth disease in children and is responsible for thousands of deaths in the past 20 years. Food azo dyes have been widely used since the nineteenth century; however, their biological effects on humans and microbes residing in humans are poorly understood. Here, we discovered that one of these dyes, brilliant black BN (E151), was particularly effective in inhibiting the infectivity of EV71 in both cell culture and mouse model studies. Mechanistic studies demonstrated that these sulfonated dyes mainly competed with EV71 attachment factors for viral binding to block viral attachment/entry to host cells. As no commercial antiviral drugs against EV71 are currently available, our findings open an avenue to exploit the development of permitted food dye E151 as a potential anti-EV71 agent.


Assuntos
Compostos Azo/farmacologia , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/tratamento farmacológico , Virulência/efeitos dos fármacos , Animais , Chlorocebus aethiops , Ciclofilina A/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Enterovirus Humano A/efeitos dos fármacos , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/virologia , Aditivos Alimentares/farmacologia , Humanos , Camundongos , Células Vero , Ligação Viral/efeitos dos fármacos
20.
Front Microbiol ; 10: 815, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31057520

RESUMO

Avian influenza A viruses (AIVs) have a broad host range, but are most intimately associated with waterfowl (Anseriformes) and, in the case of the H13 and H16 subtypes, gulls (Charadriiformes). Host associations are multifactorial, but a key factor is the ability of the virus to bind host cell receptors and thereby initiate infection. The current study aims at investigating the tissue attachment pattern of a panel of AIVs, comprising H3N2, H6N1, H12N5, and H16N3, to avian trachea and colon tissue samples obtained from host species of different orders. Virus attachment was not restricted to the bird species or order from which the virus was isolated. Instead, extensive virus attachment was observed to several distantly related avian species. In general, more virus attachment and receptor expression were observed in trachea than in colon samples. Additionally, a human seasonal H3N2 virus was studied. Unlike the studied AIVs, this virus mainly attached to tracheae from Charadriiformes and a very limited set of avian cola. In conclusion, the reported results highlight the importance of AIV attachment to trachea in many avian species. Finally, the importance of chickens and mallards in AIVs dynamics was illustrated by the abundant AIV attachment observed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA