Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
1.
Biomed Pharmacother ; 174: 116558, 2024 May.
Article in English | MEDLINE | ID: mdl-38603887

ABSTRACT

Human adenovirus (HAdV) infection is a major cause of respiratory disease, yet no antiviral drugs have been approved for its treatment. Herein, we evaluated the antiviral and anti-inflammatory effects of cyclin-dependent protein kinase (CDK) inhibitor indirubin-3'-monoxime (IM) against HAdV infection in cells and a transgenic mouse model. After evaluating its cytotoxicity, cytopathic effect reduction, antiviral replication kinetics, and viral yield reduction assays were performed to assess the anti-HAdV activity of IM. Quantitative real-time polymerase chain reaction (qPCR), quantitative reverse transcription PCR (qRT-PCR), and western blotting were used to assess the effects of IM on HAdV DNA replication, transcription, and protein expression, respectively. IM significantly inhibited HAdV DNA replication as well as E1A and Hexon transcription, in addition to significantly suppressing the phosphorylation of the RNA polymerase II C-terminal domain (CTD). IM mitigated body weight loss, reduced viral burden, and lung injury, decreasing cytokine and chemokine secretion to a greater extent than cidofovir. Altogether, IM inhibits HAdV replication by downregulating CTD phosphorylation to suppress viral infection and corresponding innate immune reactions as a promising therapeutic agent.


Subject(s)
Adenoviruses, Human , Anti-Inflammatory Agents , Antiviral Agents , Indoles , Oximes , Virus Replication , Indoles/pharmacology , Animals , Oximes/pharmacology , Humans , Antiviral Agents/pharmacology , Adenoviruses, Human/drug effects , Virus Replication/drug effects , Anti-Inflammatory Agents/pharmacology , Mice , Mice, Transgenic , Adenovirus Infections, Human/drug therapy , Adenovirus Infections, Human/virology , A549 Cells , Cytokines/metabolism , Phosphorylation/drug effects
2.
Viruses ; 16(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38675973

ABSTRACT

Differentiated HepaRG cells are popular in vitro cell models for hepatotoxicity studies. Their differentiation is usually supported by the addition of dimethyl sulfoxide (DMSO), an amphipathic solvent widely used in biomedicine, for example, in potential novel therapeutic drugs and cryopreservation of oocytes. Recent studies have demonstrated drastic effects, especially on epigenetics and extracellular matrix composition, induced by DMSO, making its postulated inert character doubtful. In this work, the influence of DMSO and DMSO-mediated modulation of differentiation on human adenovirus (HAdV) infection of HepaRG cells was investigated. We observed an increase in infectivity of HepaRG cells by HAdVs in the presence of 1% DMSO. However, this effect was dependent on the type of medium used for cell cultivation, as cells in William's E medium showed significantly stronger effects compared with those cultivated in DMEM. Using different DMSO concentrations, we proved that the impact of DMSO on infectability was dose-dependent. Infection of cells with a replication-deficient HAdV type demonstrated that the mode of action of DMSO was based on viral entry rather than on viral replication. Taken together, these results highlight the strong influence of the used cell-culture medium on the performed experiments as well as the impact of DMSO on infectivity of HepaRG cells by HAdVs. As this solvent is widely used in cell culture, those effects must be considered, especially in screening of new antiviral compounds.


Subject(s)
Adenoviruses, Human , Cell Differentiation , Dimethyl Sulfoxide , Virus Replication , Dimethyl Sulfoxide/pharmacology , Humans , Adenoviruses, Human/drug effects , Adenoviruses, Human/physiology , Cell Differentiation/drug effects , Cell Line , Virus Replication/drug effects , Virus Internalization/drug effects , Hepatocytes/virology , Hepatocytes/drug effects , Adenovirus Infections, Human/virology , Culture Media/chemistry
3.
Int J Antimicrob Agents ; 63(5): 107116, 2024 May.
Article in English | MEDLINE | ID: mdl-38401774

ABSTRACT

Human adenovirus (HAdV) and cytomegalovirus (HCMV) cause high morbidity and mortality in patients undergoing solid organ transplantation (SOT) and haematopoietic stem cell transplantation (HSCT). Immunosuppressors are used universally to prevent graft-vs-host disease in HSCT and graft rejection in SOT. The long-term use of these drugs is associated with a high risk of infection, but there is also evidence of their specific interference with viral infection. This study evaluated the antiviral activity of immunosuppressors commonly used in clinical practice in SOT and HSCT recipients in vitro to determine whether their use could be associated with reduced risk of HAdV and HCMV infection. Cyclophosphamide, tacrolimus, cyclosporine, mycophenolic acid, methotrexate, everolimus and sirolimus presented antiviral activity, with 50% inhibitory concentration (IC50) values at low micromolar and sub-micromolar concentrations. Mycophenolic acid and methotrexate showed the greatest antiviral effects against HAdV (IC50=0.05 µM and 0.3 µM, respectively) and HCMV (IC50=10.8 µM and 0.02 µM, respectively). The combination of tacrolimus and mycophenolic acid showed strong synergistic antiviral activity against both viruses, with combinatory indexes (CI50) of 0.02 and 0.25, respectively. Additionally, mycophenolic acid plus cyclosporine, and mycophenolic acid plus everolimus/sirolimus showed synergistic antiviral activity against HAdV (CI50=0.05 and 0.09, respectively), while methotrexate plus cyclosporine showed synergistic antiviral activity against HCMV (CI50=0.29). These results, showing antiviral activity in vitro against both HAdV and HCMV, at concentrations below the human Cmax values, may be relevant for the selection of specific immunosuppressant therapies in patients at risk of HAdV and HCMV infections.


Subject(s)
Adenoviruses, Human , Antiviral Agents , Cytomegalovirus , Immunosuppressive Agents , Humans , Immunosuppressive Agents/pharmacology , Antiviral Agents/pharmacology , Adenoviruses, Human/drug effects , Cytomegalovirus/drug effects , Drug Synergism , Inhibitory Concentration 50 , Mycophenolic Acid/pharmacology , Tacrolimus/pharmacology , Cyclosporine/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/prevention & control
4.
Viruses ; 13(10)2021 10 02.
Article in English | MEDLINE | ID: mdl-34696411

ABSTRACT

Viral proteases are indispensable for successful virion maturation, thus making them a prominent drug target. Their enzyme activity is tightly spatiotemporally regulated by expression in the precursor form with little or no activity, followed by activation via autoprocessing. These cleavage events are frequently triggered upon transportation to a specific compartment inside the host cell. Typically, precursor oligomerization or the presence of a co-factor is needed for activation. A detailed understanding of these mechanisms will allow ligands with non-canonical mechanisms of action to be designed, which would specifically modulate the initial irreversible steps of viral protease autoactivation. Binding sites exclusive to the precursor, including binding sites beyond the protease domain, can be exploited. Both inhibition and up-regulation of the proteolytic activity of viral proteases can be detrimental for the virus. All these possibilities are discussed using examples of medically relevant viruses including herpesviruses, adenoviruses, retroviruses, picornaviruses, caliciviruses, togaviruses, flaviviruses, and coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Viral Protease Inhibitors/pharmacology , Viral Proteases/metabolism , Virus Diseases/drug therapy , Adenoviruses, Human/drug effects , Adenoviruses, Human/metabolism , Flavivirus/drug effects , Flavivirus/metabolism , HIV-1/drug effects , Herpesviridae/drug effects , Herpesviridae/metabolism , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Viral Proteases/biosynthesis
5.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576237

ABSTRACT

Previous studies reported on the broad-spectrum antiviral function of heparin. Here we investigated the antiviral function of magnesium-modified heparin and found that modified heparin displayed a significantly enhanced antiviral function against human adenovirus (HAdV) in immortalized and primary cells. Nuclear magnetic resonance analyses revealed a conformational change of heparin when complexed with magnesium. To broadly explore this discovery, we tested the antiviral function of modified heparin against herpes simplex virus type 1 (HSV-1) and found that the replication of HSV-1 was even further decreased compared to aciclovir. Moreover, we investigated the antiviral effect against the new severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and measured a 55-fold decreased viral load in the supernatant of infected cells associated with a 38-fold decrease in virus growth. The advantage of our modified heparin is an increased antiviral effect compared to regular heparin.


Subject(s)
Antiviral Agents/pharmacology , Heparin/pharmacology , Magnesium Chloride/pharmacology , Acyclovir/pharmacology , Adenoviruses, Human/drug effects , Adenoviruses, Human/physiology , Animals , Antiviral Agents/chemistry , CHO Cells , Cell Line, Tumor , Chlorocebus aethiops , Cricetulus , Drug Evaluation, Preclinical , Fibroblasts , Heparin/chemistry , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Magnesium Chloride/chemistry , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Structure , Primary Cell Culture , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Structure-Activity Relationship , Vero Cells , Viral Load/drug effects , Virus Replication/drug effects
6.
Viruses ; 13(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34452467

ABSTRACT

Type III interferons (lambda IFNs) are a quite new, small family of three closely related cytokines with interferon-like activity. Attention to IFN-λ is mainly focused on direct antiviral activity in which, as with IFN-α, viral genome replication is inhibited without the participation of immune system cells. The heterodimeric receptor for lambda interferons is exposed mainly on epithelial cells, which limits its possible action on other cells, thus reducing the likelihood of developing undesirable side effects compared to type I IFN. In this study, we examined the antiviral potential of exogenous human IFN-λ1 in cellular models of viral infection. To study the protective effects of IFN-λ1, three administration schemes were used: 'preventive' (pretreatment); 'preventive/therapeutic' (pre/post); and 'therapeutic' (post). Three IFN-λ1 concentrations (from 10 to 500 ng/mL) were used. We have shown that human IFN-λ1 restricts SARS-CoV-2 replication in Vero cells with all three treatment schemes. In addition, we have shown a decrease in the viral loads of CHIKV and IVA with the 'preventive' and 'preventive/therapeutic' regimes. No significant antiviral effect of IFN-λ1 against AdV was detected. Our study highlights the potential for using IFN-λ as a broad-spectrum therapeutic agent against respiratory RNA viruses.


Subject(s)
Adenoviruses, Human/drug effects , Chikungunya virus/drug effects , Influenza A virus/drug effects , Interferons/pharmacology , SARS-CoV-2/drug effects , A549 Cells , Adenoviruses, Human/physiology , Animals , Chikungunya virus/physiology , Chlorocebus aethiops , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Influenza A virus/physiology , Interferons/therapeutic use , Interleukins , RNA Virus Infections/drug therapy , RNA Virus Infections/prevention & control , Recombinant Proteins/pharmacology , SARS-CoV-2/physiology , Vero Cells , Viral Load/drug effects , Virus Replication/drug effects , Interferon Lambda
7.
Molecules ; 26(11)2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34198721

ABSTRACT

There is no approved antiviral therapy for adenovirus (HAdV) ocular infections. Astodrimer sodium (SPL7013) is a polyanionic dendrimer with antiviral activity. The current study evaluated the ocular tolerability and anti-adenoviral efficacy of topical SPL7013 in rabbit ocular models. In a tolerability study, rabbits were treated with 3% SPL7013, vehicle, or 0.5% cidofovir. Their eyes were graded using the Draize scale. In antiviral efficacy studies, HAdV5 inoculated eyes were treated with 3% SPL7013, vehicle, or 0.5% cidofovir. Eyes were cultured for the virus on days 0, 1, 3, 4, 5, 7, 9, 11, and 14. Viral titers were determined. There were no differences in Draize scores between 3% SPL7013 and vehicle on any day. Cidofovir produced significantly higher Draize scores on day 12 than SPL7013 and vehicle. The 3% SPL7013 and 0.5% cidofovir significantly reduced daily viral titers and positive cultures per total compared with vehicle on several different days. The 3% SPL7013 and 0.5% cidofovir significantly reduced the duration of HAdV5 shedding compared to vehicle. The 3% SPL7013 demonstrated significantly more antiviral activity compared with vehicle in the Ad5/NZW rabbit ocular model. The 3% SPL7013 induced "minimal" to "practically non-irritating" Draize scores in the ocular tolerability study. Further development of astodrimer sodium as a topical antiviral therapy for adenoviral ocular infections is indicated.


Subject(s)
Adenoviridae Infections/drug therapy , Cidofovir/administration & dosage , Dendrimers/administration & dosage , Eye Infections, Viral/drug therapy , Polylysine/administration & dosage , A549 Cells , Adenoviruses, Human/drug effects , Adenoviruses, Human/physiology , Administration, Topical , Animals , Cidofovir/pharmacology , Dendrimers/pharmacology , Disease Models, Animal , Female , Humans , Polylysine/pharmacology , Rabbits , Treatment Outcome , Viral Load/drug effects
8.
Antiviral Res ; 188: 105034, 2021 04.
Article in English | MEDLINE | ID: mdl-33577808

ABSTRACT

Human adenoviruses (HAdV) are ubiquitous human pathogens that cause a significant burden of respiratory, ocular, and gastrointestinal illnesses. Although HAdV infections are generally self-limiting, pediatric and immunocompromised individuals are at particular risk for developing severe disease. Currently, no approved antiviral therapies specific to HAdV exist. Recent outbreaks underscore the need for effective antiviral agents to treat life-threatening infections. In this review we will focus on recent developments in search of potential therapeutic agents for controlling HAdV infections, with a focus on those targeting post-entry stages of the virus replicative cycle.


Subject(s)
Adenovirus Infections, Human/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/therapeutic use , Active Transport, Cell Nucleus/drug effects , Adenovirus Infections, Human/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Antiviral Agents/pharmacology , DNA Replication/drug effects , Drug Repositioning , Drug Therapy, Combination , Epigenesis, Genetic/drug effects , Gene Expression/drug effects , Humans , Immunotherapy, Adoptive , T-Lymphocytes/immunology , Virus Replication/drug effects
9.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562748

ABSTRACT

Human adenoviruses (HAdVs) display a wide range of tissue tropism and can cause an array of symptoms from mild respiratory illnesses to disseminated and life-threatening infections in immunocompromised individuals. However, no antiviral drug has been approved specifically for the treatment of HAdV infections. Herein, we report our continued efforts to optimize salicylamide derivatives and discover compound 16 (JMX0493) as a potent inhibitor of HAdV infection. Compound 16 displays submicromolar IC50 values, a higher selectivity index (SI > 100) and 2.5-fold virus yield reduction compared to our hit compound niclosamide. Moreover, unlike niclosamide, our mechanistic studies suggest that the antiviral activity of compound 16 against HAdV is achieved through the inhibition of viral particle escape from the endosome, which bars subsequent uncoating and the presentation of lytic protein VI.


Subject(s)
Adenoviruses, Human/physiology , Antiviral Agents/pharmacology , Endosomes/virology , Niclosamide/pharmacology , Salicylamides/pharmacology , A549 Cells , Adenoviruses, Human/drug effects , Drug Discovery , Endosomes/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Niclosamide/chemistry , Salicylamides/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Tropism , Virus Internalization/drug effects , Virus Replication/drug effects
10.
Virology ; 555: 102-110, 2021 03.
Article in English | MEDLINE | ID: mdl-33032802

ABSTRACT

Human adenovirus (HAdV) can cause severe disease in certain at-risk populations such as newborns, young children, the elderly and individuals with a compromised immune system. Unfortunately, no FDA-approved antiviraldrug is currently available for the treatment of HAdV infections. Within the nucleus of infected cells, the HAdV genome associates with histones and forms a chromatin-like structure during early infection, and viral gene expression appears to be regulated by cellular epigenetic processes. Thus, one potential therapeutic strategy to combat HAdV disease may be to target the cellular proteins involved in modifying the viral nucleoprotein structure and facilitating HAdV gene expression and replication. We have screened a panel of small molecules that modulate the activity of epigenetic regulatory proteins for compounds affecting HAdV gene expression. Several of the compounds, specifically chaetocin, gemcitabine and lestaurtinib, reduced HAdV recovery by 100- to 1000-fold, while showing limited effects on cell health, suggesting that these compounds may indeed be promising as anti-HAdV therapeutics.


Subject(s)
Adenovirus Infections, Human/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Virus Replication/drug effects , Carbazoles/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Furans/pharmacology , Humans , Piperazines/pharmacology , Gemcitabine
11.
Eur J Ophthalmol ; 31(2): 379-384, 2021 Mar.
Article in English | MEDLINE | ID: mdl-31813297

ABSTRACT

PURPOSE: The aim of this study was to test the antiviral effectivity of potassium peroxymonosulfate (RUBYSTA®, KYORIN) against five epidemic keratoconjunctivitis-related types of Human adenovirus D in vitro. METHODS: Five types of Human adenovirus D (8, 37, 53, 54 and 56) were incubated with 1% potassium peroxymonosulfate, 0.1% sodium hypochlorite (NaClO) or alcohol-based disinfectant for 30 s or 1 min. These solutions were subjected to measurements of viral titres by infection assays in A549 cells. At day 6 post-infection, both, supernatants and cells, were collected and the viral genome was assessed by real-time polymerase chain reaction analysis. RESULTS: Treatments with 1% potassium peroxymonosulfate led to significant reduction in all tested Human adenovirus D types comparable to disinfecting effects by 0.1% NaClO. Overall, potassium peroxymonosulfate demonstrated sufficient inactivation of the major epidemic keratoconjunctivitis-causing Human adenovirus D to be considered for disinfection and prevention purposes in ophthalmological clinics and hospitals. CONCLUSION: This study demonstrated that potassium peroxymonosulfate is a promising disinfectant for the prevention of epidemic keratoconjunctivitis nosocomial infections in ophthalmological clinics.


Subject(s)
Adenovirus Infections, Human/virology , Adenoviruses, Human/drug effects , Disinfectants/pharmacology , Keratoconjunctivitis/virology , Oxidants/pharmacology , Peroxides/pharmacology , A549 Cells , Cross Infection/prevention & control , Epidemics , Humans , Virus Replication/drug effects
12.
J Med Chem ; 63(21): 12830-12852, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33112138

ABSTRACT

An effective therapy for human adenovirus (HAdV) infections in immunocompromised patients and healthy individuals with community-acquired pneumonia remains an unmet medical need. We herein reported a series of novel substituted N-(4-amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogues as potent HAdV inhibitors. Compounds 6, 15, 29, 40, 43, 46, 47, and 54 exhibited increased selectivity indexes (SI > 100) compared to the lead compound niclosamide, while maintaining sub-micromolar to low micromolar potency against HAdV. The preliminary mechanistic studies indicated that compounds 6 and 43 possibly target the HAdV DNA replication process, while compounds 46 and 47 suppress later steps of HAdV life cycle. Notably, among these derivatives, compound 15 showed improved anti-HAdV activity (IC50 = 0.27 µM), significantly decreased cytotoxicity (CC50 = 156.8 µM), and low in vivo toxicity (maximum tolerated dose = 150 mg/kg in hamster) as compared with niclosamide, supporting its further in vivo efficacy studies for the treatment of HAdV infections.


Subject(s)
Adenoviruses, Human/physiology , Antiviral Agents/chemistry , Benzamides/chemistry , Adenoviruses, Human/drug effects , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Drug Evaluation, Preclinical , Humans , Lethal Dose 50 , Structure-Activity Relationship , Virus Internalization/drug effects , Virus Replication/drug effects
13.
Sci Data ; 7(1): 265, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32788590

ABSTRACT

Human adenoviruses (HAdVs) are fatal to immuno-suppressed individuals, but no effective anti-HAdV therapy is available. Here, we present a novel image-based high-throughput screening (HTS) platform, which scores the full viral replication cycle from virus entry to dissemination of progeny and second-round infections. We analysed 1,280 small molecular weight compounds of the Prestwick Chemical Library (PCL) for interference with HAdV-C2 infection in a quadruplicate, blinded format, and performed robust image analyses and hit filtering. We present the entire set of the screening data including all images, image analyses and data processing pipelines. The data are made available at the Image Data Resource (IDR, idr0081). Our screen identified Nelfinavir mesylate as an inhibitor of HAdV-C2 multi-round plaque formation, but not single round infection. Nelfinavir has been FDA-approved for anti-retroviral therapy in humans. Our results underscore the power of image-based full cycle infection assays in identifying viral inhibitors with clinical potential.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Small Molecule Libraries/pharmacology , Adenoviruses, Human/physiology , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Nelfinavir/pharmacology , Virus Replication/drug effects
14.
Antiviral Res ; 182: 104872, 2020 10.
Article in English | MEDLINE | ID: mdl-32768412

ABSTRACT

OBJECTIVES: Human adenovirus (HAdV) infections are associated with a high morbidity and mortality in transplant patients requiring the use of antiviral treatments. Brincidofovir (BCV), a cytidine analog, inhibits HAdV replication through viral DNA elongation termination and likely through other mechanisms. To elucidate if BCV regulates cellular antiviral pathways, we analyzed its impact on HAdV-infected and non-HAdV-infected lung epithelial cells. METHODS: We assessed the cellular and viral transcriptome of A549 cells infected and non-infected with HAdV C5 and treated or non-treated with BCV by RNAseq after 72 h. RESULTS: BCV treatment of HAdV infected cells resulted in a profound decrease of viral transcription associated with a relative overexpression of the early genes E1A and E4 and of the late gene L1. BCV had also a profound impact on A549 cells' transcriptome. Ontologic analysis revealed an effect of BCV on several pathways known to interact with adenovirus replication as mTor signalling and Wnt pathways. A549 cells treated with BCV demonstrated a significant inhibition of the biological function of "viral replication" including 25 dysregulated genes involved in inflammation pathways. CONCLUSION: We demonstrated that BCV alters viral gene expression and promotes the expression of antiviral cellular pathways in A549 cells. These results provide new insights how to interfere with cellular pathways to control HAdV infections.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Organophosphonates/pharmacology , Transcriptome , A549 Cells , Cytosine/pharmacology , Host Microbial Interactions , Humans , Virus Replication/drug effects
15.
ACS Chem Biol ; 15(10): 2683-2691, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32845119

ABSTRACT

Coxsackievirus A24 variant (CVA24v) and human adenovirus 37 (HAdV-37) are leading causative agents of the severe and highly contagious ocular infections acute hemorrhagic conjunctivitis and epidemic keratoconjunctivitis, respectively. Currently, neither vaccines nor antiviral agents are available for treating these diseases, which affect millions of individuals worldwide. CVA24v and HAdV-37 utilize sialic acid as attachment receptors facilitating entry into host cells. Previously, we and others have shown that derivatives based on sialic acid are effective in preventing HAdV-37 binding and infection of cells. Here, we designed and synthesized novel pentavalent sialic acid conjugates and studied their inhibitory effect against CVA24v and HAdV-37 binding and infection of human corneal epithelial cells. The pentavalent conjugates are the first reported inhibitors of CVA24v infection and proved efficient in blocking HAdV-37 binding. Taken together, the pentavalent conjugates presented here form a basis for the development of general inhibitors of these highly contagious ocular pathogens.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Enterovirus C, Human/drug effects , Sialic Acids/pharmacology , Adenoviruses, Human/chemistry , Binding Sites , Enterovirus C, Human/chemistry , Humans , Virus Attachment/drug effects , Virus Internalization/drug effects
16.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641484

ABSTRACT

Human adenoviruses (HAdV) are ubiquitous within the human population and comprise a significant burden of respiratory illnesses worldwide. Pediatric and immunocompromised individuals are at particular risk for developing severe disease; however, no approved antiviral therapies specific to HAdV exist. Ivermectin is an FDA-approved broad-spectrum antiparasitic drug that also exhibits antiviral properties against a diverse range of viruses. Its proposed function is inhibiting the classical protein nuclear import pathway mediated by importin-α (Imp-α) and -ß1 (Imp-ß1). Many viruses, including HAdV, rely on this host pathway for transport of viral proteins across the nuclear envelope. In this study, we show that ivermectin inhibits HAdV-C5 early gene transcription, early and late protein expression, genome replication, and production of infectious viral progeny. Similarly, ivermectin inhibits genome replication of HAdV-B3, a clinically important pathogen responsible for numerous recent outbreaks. Mechanistically, we show that ivermectin disrupts binding of the viral E1A protein to Imp-α without affecting the interaction between Imp-α and Imp-ß1. Our results further extend ivermectin's broad antiviral activity and provide a mechanistic underpinning for its mode of action as an inhibitor of cellular Imp-α/ß1-mediated nuclear import.IMPORTANCE Human adenoviruses (HAdVs) represent a ubiquitous and clinically important pathogen without an effective antiviral treatment. HAdV infections typically cause mild symptoms; however, individuals such as children, those with underlying conditions, and those with compromised immune systems can develop severe disseminated disease. Our results demonstrate that ivermectin, an FDA-approved antiparasitic agent, is effective at inhibiting replication of several HAdV types in vitro This is in agreement with the growing body of literature suggesting ivermectin has broad antiviral activity. This study expands our mechanistic knowledge of ivermectin by showing that ivermectin targets the ability of importin-α (Imp-α) to recognize nuclear localization sequences, without effecting the Imp-α/ß1 interaction. These data also exemplify the applicability of targeting host factors upon which viruses rely as a viable antiviral strategy.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Adenoviruses, Human/drug effects , Antiparasitic Agents/pharmacology , Ivermectin/pharmacology , Virus Replication/drug effects , alpha Karyopherins/genetics , beta Karyopherins/genetics , A549 Cells , Active Transport, Cell Nucleus/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Adenoviruses, Human/pathogenicity , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/virology , Cytosol/drug effects , Cytosol/metabolism , Cytosol/virology , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Signal Transduction , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/metabolism , beta Karyopherins/metabolism
17.
Viruses ; 12(6)2020 06 18.
Article in English | MEDLINE | ID: mdl-32570859

ABSTRACT

The new epidemiological scenario has so far focused on the environmental circulation of human viral pathogens. Owing to the side effects of chemical disinfectants, there is an increasing need for knowledge on the use of virucidal compounds, especially those of a natural origin. Climacostol is a molecule produced by a freshwater ciliate and it exhibits activity against bacterial and fungal pathogens. We thus also speculated that there might be an effect on viral viability, which has never been tested. To evaluate such activity, we chose human adenovirus (HAdV), which is representative of waterborne viruses. We conducted experiments using HAdV serotype 5, whose titer was determined by infecting HeLa cell cultures. HAdV5 was shown to be sensitive to climacostol at a concentration of 0.0002 mg/mL, with an approximate 3 Log10 reduction when the initial titer of HAdV5 was approximately 104 and 103 TCID50/mL. These preliminary results could be an important starting point for further research aimed at improving the characterization of climacostol activity under different experimental conditions and against various viruses, including enveloped ones (i.e., the coronavirus). The production of climacostol by a protist living in fresh water also suggests a possible application in the activated sludge of wastewater treatment plants.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Disinfectants/pharmacology , Resorcinols/pharmacology , Cell Line , Ciliophora/metabolism , HeLa Cells , Humans , Preliminary Data , Sewage/virology , Water Purification/methods
18.
Int J Antimicrob Agents ; 56(2): 106046, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32540429

ABSTRACT

Human adenovirus (HAdV) infection has an important clinical impact in the immunosuppressed population and is associated with high morbidity and mortality rates. The lack of a specific, safe and effective antiviral treatment against HAdV makes necessary the search for new therapeutic options. The aim of this study was to evaluate the in vitro activity of ganciclovir (GCV) against HAdV in co-infection by human cytomegalovirus (HCMV) and HAdV in cellular cultures. Quantitative real-time polymerase chain reaction (qPCR) was used to measure HAdV and HCMV DNA replication efficiency in monocultures and in co-infection situations in the presence of both cidofovir (CDV) and GCV. The effects of GCV and CDV were also evaluated in a burst assay (used to measure the production of virus particles) for both viruses, alone and in combination. GCV decreased by 1-log the HAdV DNA replication efficiency in co-infection with HCMV compared with its activity in HAdV monoculture. The burst assay showed that the reductions in virus yield in the presence of GCV were higher for HCMV and co-infection than for HAdV in monoculture (145.2±35.5- vs. 116.4±27.3- vs. 23.0±10.0-fold, respectively, P<0.05). The improved anti-HAdV activity of GCV during co-infection may be because of the more efficient phosphorylation of GCV by the HCMV protein kinase, UL97. Patients treated with GCV as pre-emptive therapy for HCMV infection may be considered as low-risk for developing HAdV infections; however, further evaluations are required to confirm these results.


Subject(s)
Adenoviruses, Human/drug effects , Cidofovir/pharmacology , Cytomegalovirus/drug effects , Ganciclovir/pharmacology , A549 Cells , Adenovirus Infections, Human/drug therapy , Adenovirus Infections, Human/virology , Adenoviruses, Human/genetics , Antiviral Agents/pharmacology , Cell Line , Coinfection/drug therapy , Coinfection/virology , Cytomegalovirus/genetics , DNA, Viral/metabolism , Humans , Inhibitory Concentration 50 , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry
19.
Antiviral Res ; 179: 104789, 2020 07.
Article in English | MEDLINE | ID: mdl-32353383

ABSTRACT

Human adenoviruses (HAdVs) often cause mild respiratory infections. These infections, however, can potentially become fatal in immunosuppressive patients. Unfortunately, there has been no specific anti-HAdV drug approved for treatment of HAdV infections. In this study, a time-course transcriptome of HAdV-infected human lung epithelial cells (A549 cells) was performed and compared with perturbation datasets of 890 drug-treated A549 cells from the library of integrated network-based cellular signatures (LINCS) database to predict previously unknown therapeutic drug-HAdV relationships using a characteristic direction (CD) algorithm. We performed experiments to validate a prediction for the anti-diabetic drug rosiglitazone as a candidate drug for treatment of anti-HAdV both in vivo and in vitro. The Type I interferon (IFNs) signaling pathway was negatively regulated during the course of HAdV infection and rosiglitazone increased STAT1 phosphorylation for antiviral IFN response induction. Taken together, this study confirmed the prospect for re-exploitation of this FDA-approved drug as a potential therapeutic for HAdV infections.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/therapeutic use , Databases, Pharmaceutical , Drug Repositioning , Rosiglitazone/therapeutic use , Virus Replication/drug effects , A549 Cells , Adenovirus Infections, Human/drug therapy , Adenovirus Infections, Human/virology , Adenoviruses, Human/physiology , Algorithms , Animals , Antiviral Agents/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Female , Humans , Immunocompromised Host , Mice , Mice, SCID , Rosiglitazone/pharmacology
20.
Antivir Chem Chemother ; 28: 2040206620921319, 2020.
Article in English | MEDLINE | ID: mdl-32345035

ABSTRACT

Human adenovirus type 19 (HAdV-19) is a major cause of the epidemic keratoconjunctivitis. Outbreaks of keratoconjunctivitis are problematic to human health, especially for infants, the elderly, and immunocompromised individuals. However, the development of anti-HAdV drugs has been hampered by inconvenient screening systems; therefore, development of a simple screening method is highly desirable. In this study, we identified that HAdV-19 can infect a human lymphoid cell line transformed with human T-cell leukemia virus (MT-2 cells). MT-2 cells supported HAdV-19 replication and showed apparent cytopathic effects within five days post-infection. Using a thiazolyl blue tetrazolium bromide (MTT)-based colorimetric assay on MT-2 cells, we were able to detect the anti-HAdV-19 activities of previously reported nucleoside/tide compounds, including (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine (cidofovir), 2',3'-dideoxycytidine (zalcitabine) and 3'-deoxy-3'-fluorothymidine (trifluridine). Compared with previous methods, this system represents a more simple and rapid method to screen anti-HAdV-19 agents.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Cidofovir/pharmacology , Lymphocytes/drug effects , Zalcitabine/pharmacology , Adenoviruses, Human/genetics , Cells, Cultured , Humans , Keratoconjunctivitis/drug therapy , Keratoconjunctivitis/virology , Lymphocytes/virology , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...