Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Expert Opin Drug Discov ; 18(4): 371-383, 2023 04.
Article in English | MEDLINE | ID: mdl-36995192

ABSTRACT

INTRODUCTION: Flaviviruses are emerging or reemerging pathogens that have caused several outbreaks throughout the world and pose serious threats on human health and economic development. RNA-based therapeutics are developing rapidly, and hold promise in the fight against flaviviruses. However, to develop efficient and safe therapeutics for flaviviruses, many challenges remain unsolved. AREAS COVERED: In this review, the authors briefly introduced the biology of flaviviruses and the current advances in RNA-based therapeutics for them. Furthermore, the authors list the challenges and possible solutions in this area. Finally, the authors give their opinion on the development and future of RNA-based therapeutics for flaviviruses. EXPERT OPINION: With the rapid development of structural biology, the crystal structures of flavivirus proteins may lay the foundation for future rational drug design. Studies regarding the interactions between the flavivirus and the host will also be invaluable to inhibitor design. Researchers should maintain the current momentum to bring about safe and effective anti-flavivirus drugs to licensure through joint efforts of academia, government, and industry.


Subject(s)
Flavivirus Infections , Flavivirus , Humans , Flavivirus/genetics , Flavivirus/metabolism , RNA/metabolism , RNA/pharmacology , Flavivirus Infections/drug therapy
2.
Curr Med Chem ; 30(21): 2396-2420, 2023.
Article in English | MEDLINE | ID: mdl-35702779

ABSTRACT

Sulfur and oxygen containing-compounds are a relevant class of derivatives that is constantly growing due to their wide range of pharmacological activity, including the antiviral one. As proof of this, there are several FDA approved antiviral compounds having sulfur and oxygen in their structures. Among RNA viruses, the flavivirus genus (e.g. Dengue, West Nile, Yellow Fever and Zika viruses) holds a relevant place within zoonotic pathogens and thus flavivirus infections are considered a growing risk for the public health. As a consequence, the drug discovery process aimed at identify new anti- flavivirus agents is of great relevance and will help to find effective therapies not available yet. One of the most alarming features of flaviviruses is their ability to co-infect the host, thus aggravating the symptoms of the disease. Therefore, finding compounds endowed with a broad-spectrum anti-flavivirus activity is now becoming a pressing need. In this review, we describe the most promising compounds having both sulfur and oxygen in their structures characterized by a broad-spectrum activity against different flaviviruses. Furthermore, the synthetic procedures applied for the preparation of the described derivatives are also reported. Readers can be inspired by the contents of this review to design and synthesize more effective anti-flavivirus agents as well as to select viral or host targets to achieve an antiviral activity as broadly as possible.


Subject(s)
Flavivirus Infections , Flavivirus , Zika Virus Infection , Zika Virus , Humans , Flavivirus/genetics , Oxygen/therapeutic use , Flavivirus Infections/drug therapy , Zika Virus/genetics , Sulfur/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Zika Virus Infection/drug therapy
3.
Viruses ; 14(8)2022 07 27.
Article in English | MEDLINE | ID: mdl-36016263

ABSTRACT

Usutu virus (USUV) is an emerging mosquito-borne arbovirus within the genus Flavivirus, family Flaviviridae. Similar to the closely related West Nile virus (WNV), USUV infections are capable of causing mass mortality in wild and captive birds, especially blackbirds. In the last few years, a massive spread of USUV was present in the avian population of Germany and other European countries. To date, no specific antiviral therapies are available. Nine different approved drugs were tested for their antiviral effects on the replication of USUV in vitro in a screening assay. Ivermectin was identified as a potent inhibitor of USUV replication in three cell types from different species, such as simian Vero CCL-81, human A549 and avian TME R. A 2- to 7-log10 reduction of the viral titer in the supernatant was detected at a non-cytotoxic concentration of 5 µM ivermectin dependent on the applied cell line. IC50 values of ivermectin against USUV lineage Africa 3 was found to be 0.55 µM in Vero CCL-81, 1.94 µM in A549 and 1.38 µM in TME-R cells. The antiviral efficacy was comparable between the USUV lineages Africa 2, Africa 3 and Europe 3. These findings show that ivermectin may be a candidate for further experimental and clinical studies addressing the treatment of USUV disease, especially in captive birds.


Subject(s)
Bird Diseases , Flavivirus Infections , Flavivirus , Animals , Antiviral Agents/pharmacology , Birds , Flavivirus Infections/drug therapy , Flavivirus Infections/epidemiology , Flavivirus Infections/veterinary , Humans , Ivermectin/pharmacology
4.
Viruses ; 14(6)2022 06 05.
Article in English | MEDLINE | ID: mdl-35746697

ABSTRACT

Mosquito-borne flavivirus infections affect approximately 400 million people worldwide each year and are global threats to public health. The common diseases caused by such flaviviruses include West Nile, yellow fever, dengue, Zika infection and Japanese encephalitis, which may result in severe symptoms and disorders of multiple organs or even fatal outcomes. Till now, no specific antiviral agents are commercially available for the treatment of the diseases. Numerous strategies have been adopted to develop novel and promising inhibitors against mosquito-borne flaviviruses, including drugs targeting the critical viral components or essential host factors during infection. Research advances in antiflaviviral therapy might optimize and widen the treatment options for flavivirus infection. This review summarizes the current developmental progresses and involved molecular mechanisms of antiviral agents against mosquito-borne flaviviruses.


Subject(s)
Culicidae , Flavivirus Infections , Flavivirus , Zika Virus Infection , Zika Virus , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Flavivirus Infections/drug therapy , Humans
5.
Virol Sin ; 37(1): 94-106, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35234626

ABSTRACT

Flaviviruses are important arthropod-borne pathogens that represent an immense global health problem. Their unprecedented epidemic rate and unpredictable clinical features underscore an urgent need for antiviral interventions. Dehydroepiandrosterone (DHEA) is a natural occurring adrenal-derived steroid in the human body that has been associated in protection against various infections. In the present study, the plaque assay based primary screening was conducted on 32 synthetic derivatives of DHEA against Japanese encephalitis virus (JEV) to identify potent anti-flaviviral compounds. Based on primary screening, HAAS-AV3026 and HAAS-AV3027 were selected as hits from DHEA derivatives that exhibited strong antiviral activity against JEV (IC50 â€‹= â€‹2.13 and 1.98 â€‹µmol/L, respectively) and Zika virus (ZIKV) (IC50 â€‹= â€‹3.73 and 3.42 â€‹µmol/L, respectively). Mechanism study indicates that HAAS-AV3026 and HAAS-AV3027 do not exhibit inhibitory effect on flavivirus binding and entry process, while significantly inhibit flavivirus infection at the replication stage. Moreover, indirect immunofluorescence assay, Western blot analyses, and quantitative reverse transcription-PCR (qRT-PCR) revealed a potent antiviral activity of DHEA derivatives hits against JEV and ZIKV in terms of inhibition of viral infection, protein production, and viral RNA synthesis in Vero cells. Taken together, our results may provide a basis for the development of new antivirals against flaviviruses.


Subject(s)
Flavivirus Infections , Flavivirus , Zika Virus Infection , Zika Virus , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Dehydroepiandrosterone/pharmacology , Flavivirus Infections/drug therapy , Humans , Vero Cells , Virus Replication
6.
Curr Opin Virol ; 49: 164-175, 2021 08.
Article in English | MEDLINE | ID: mdl-34171540

ABSTRACT

The flavivirus are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than 70 viruses, and despite genomic and structural similarities, infections by different flaviviruses result in different clinical presentations. In the absence of a safe and effective vaccine against these infections, the search for new strategies to inhibit viral infection is necessary. The life cycle of arboviruses begins with the entry process composed of multiple steps: attachment, internalization, endosomal escape and capsid uncoating. This mini-review describes factors and mechanisms involved in the viral entry as events required to take over the cellular machinery and host factors and cellular pathways commonly used by flaviviruses as possible approaches for developing broad-spectrum antiviral drugs.


Subject(s)
Flavivirus Infections/virology , Flavivirus/physiology , Virus Internalization , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Endocytosis , Flavivirus/drug effects , Flavivirus/pathogenicity , Flavivirus Infections/drug therapy , Host-Pathogen Interactions , Humans , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Virus Replication
7.
Front Immunol ; 12: 630571, 2021.
Article in English | MEDLINE | ID: mdl-33968023

ABSTRACT

Tetraspanin family of proteins participates in numerous fundamental signaling pathways involved in viral transmission, virus-specific immunity, and virus-mediated vesicular trafficking. Studies in the identification of novel therapeutic candidates and strategies to target West Nile virus, dengue and Zika viruses are highly warranted due to the failure in development of vaccines. Recent evidences have shown that the widely distributed tetraspanin proteins may provide a platform for the development of novel therapeutic approaches. In this review, we discuss the diversified and important functions of tetraspanins in exosome/extracellular vesicle biology, virus-host interactions, virus-mediated vesicular trafficking, modulation of immune mechanism(s), and their possible role(s) in host antiviral defense mechanism(s) through interactions with noncoding RNAs. We also highlight the role of tetraspanins in the development of novel therapeutics to target arthropod-borne flaviviral diseases.


Subject(s)
Flavivirus Infections/drug therapy , Host Microbial Interactions/physiology , Tetraspanins/physiology , Exosomes/physiology , Extracellular Vesicles/physiology , Humans , Immunomodulation , RNA, Untranslated/physiology
8.
J Vet Med Sci ; 83(4): 734-741, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33716232

ABSTRACT

Duck Tembusu virus (DTMUV), a neurotropic flavivirus, is a causative agent of severe neurological diseases in different birds. No approved vaccines or antiviral therapeutic treatments are available to date. The poultry industry experiences significant economic losses due to DTMUV infections. Minocycline is a second-generation semi-synthetic tetracycline analogue that is commonly used as an antimicrobial treatment. Experimental studies have indicated the successful protective effects of minocycline against neuronal cell death from neurodegenerative diseases and viral encephalitis. The aim of this study was to investigate the effects of minocycline on DTMUV infection in neurons. Primary duck neurons were treated with minocycline, which exhibited neuroprotective effects via anti-apoptotic function rather than through viral replication inhibition. Minocycline might serve as a potential effective drug in DTMUV infection.


Subject(s)
Flavivirus Infections , Flavivirus , Pharmaceutical Preparations , Poultry Diseases , Animals , Ducks , Flavivirus Infections/drug therapy , Flavivirus Infections/prevention & control , Flavivirus Infections/veterinary , Minocycline/pharmacology , Neurons , Poultry Diseases/drug therapy , Poultry Diseases/prevention & control
9.
Poult Sci ; 100(4): 100989, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33647721

ABSTRACT

The duck Tembusu virus (DTMUV) is a novel mosquito-borne Flavivirus which caused huge economic losses for poultry industries in Southeast Asia and China. Currently, no effective antiviral drugs against this virus have been reported. (-)-Epigallocatechin-3-gallate (EGCG), a polyphenol present in abundance in green tea, has recently been demonstrated to have an antiviral activity for many viruses; however, whether EGCG can inhibit DTMUV infection remains unknown. Here, we tried to explore the anti-DTMUV effects and mechanisms of EGCG both in vitro and in vivo. Several EGCG treatment regimens were used to study the comprehensive antiviral activity of EGCG in DTMUV-infected baby hamster kidney cell line (BHK-21). The DTMUV titers of mock- and EGCG-treated infected cell cultures were determined using the tissue culture infective dose assay and the DTMUV mRNA copy number as determined using quantitative Real Time PCR. Moreover, the therapeutic efficacy of EGCG against DTMUV was assessed in DTMUV-infected ducklings. Our results suggested that EGCG significantly reduced the viral infection in BHK-21 cells in a dose-dependent manner, as reflected by the reduction of virus titers, virus copy number, and the expressions of viral E protein. We also observed that EGCG exhibited direct virucidal abilities against DTMUV. Notably, a significant reduction in virus binding ability was also observed, indicating that EGCG possesses excellent inhibitory effects on the viral adsorption step. In addition, DTMUV replication was also suppressed in BHK-21 cells treated with EGCG after viral entry, likely because of upregulation of the levels of interferon alfa and interferon beta. Finally, we also proved that EGCG exhibited anti-DTMUV efficacy in a duckling infection model because the survival rate was significantly improved. This is the first study to demonstrate the protective effect of EGCG against DTMUV, suggesting its potential use as an antiviral drug for DTMUV infection.


Subject(s)
Catechin/analogs & derivatives , Flavivirus Infections , Flavivirus , Interferon Type I , Poultry Diseases , Virus Internalization , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Catechin/pharmacology , Cell Line , China , Cricetinae , Ducks , Flavivirus/drug effects , Flavivirus Infections/drug therapy , Flavivirus Infections/veterinary , Interferon Type I/genetics , Poultry Diseases/drug therapy , Survival Analysis , Treatment Outcome , Up-Regulation/drug effects , Virus Internalization/drug effects
10.
ChemMedChem ; 15(24): 2391-2419, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32961008

ABSTRACT

Infections by flaviviruses, such as Dengue, West Nile, Yellow Fever and Zika viruses, represent a growing risk for global health. There are vaccines only for few flaviviruses while no effective treatments are available. Flaviviruses share epidemiological, structural, and ecologic features and often different viruses can co-infect the same host. Therefore, the identification of broad-spectrum inhibitors is highly desirable either for known flaviviruses or for viruses that likely will emerge in the future. Strategies targeting both virus and host factors have been pursued to identify broad-spectrum antiflaviviral agents. In this review, we describe the most promising and best characterized targets and their relative broad-spectrum inhibitors, identified by drug repurposing/libraries screenings and by focused medicinal chemistry campaigns. Finally, we discuss about future strategies to identify new broad-spectrum antiflavivirus agents.


Subject(s)
Antiviral Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Flavivirus Infections/drug therapy , Flavivirus/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Cell Line, Tumor , Chemistry, Pharmaceutical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Flavivirus/chemistry , Flavivirus/enzymology , Humans
11.
Article in English | MEDLINE | ID: mdl-32351903

ABSTRACT

Duck tembusu virus (DTMUV) is newly emerged in poultry and causes great losses to the breeding industry in China and neighboring countries. Effective antiviral strategies are still being studied. Autophagy is a cellular degradative pathway, and our lab's previous data show that autophagy promotes DTMUV replication in vitro. To study the role of autophagy further in vivo, we utilized ducks as the animal model to investigate the autophagy responses in DTMUV-targeted tissues. And also, we utilized autophagy regulators, including Rapamycin (Rapa) as the autophagy enhancer, 3-Methyladenine (3-MA) and Chloroquine (CQ) as the autophagy inhibitors, to adjust the host autophagic levels and then study the effects of autophagy on tissue damages and virus replication. As a result, we first found DTMUV infection trigged autophagy and autophagy regulator treatments regulated autophagy levels successfully in duck spleens and brains. Next, we found that autophagy inhibitors inhibited DTMUV replication and alleviated DTMUV-induced pathological symptoms, whereas the autophagy inducer treatment led to the opposite effects. And we also found that autophagic regulation was correlated with the expression of innate immune genes, including pattern recognition receptors, type I interferons, and cytokines, and caused different effects in different tissues. In summary, we demonstrated that autophagy facilitated DTMUV replication, aggravated the developments of pathological symptoms and possibly counteracts the host's innate immunity response in vivo.


Subject(s)
Flavivirus Infections , Poultry Diseases , Animals , Autophagy , China , Ducks , Flavivirus , Flavivirus Infections/drug therapy , Flavivirus Infections/veterinary , Poultry Diseases/drug therapy
12.
Antiviral Res ; 177: 104760, 2020 05.
Article in English | MEDLINE | ID: mdl-32135219

ABSTRACT

Infection by RNA viruses such as human immunodeficiency virus (HIV)-1, influenza, and dengue virus (DENV) represent a major burden for human health worldwide. Although RNA viruses replicate in the infected host cell cytoplasm, the nucleus is central to key stages of the infectious cycle of HIV-1 and influenza, and an important target of DENV nonstructural protein 5 (NS5) in limiting the host antiviral response. We previously identified the small molecule ivermectin as an inhibitor of HIV-1 integrase nuclear entry, subsequently showing ivermectin could inhibit DENV NS5 nuclear import, as well as limit infection by viruses such as HIV-1 and DENV. We show here that ivermectin's broad spectrum antiviral activity relates to its ability to target the host importin (IMP) α/ß1 nuclear transport proteins responsible for nuclear entry of cargoes such as integrase and NS5. We establish for the first time that ivermectin can dissociate the preformed IMPα/ß1 heterodimer, as well as prevent its formation, through binding to the IMPα armadillo (ARM) repeat domain to impact IMPα thermal stability and α-helicity. We show that ivermectin inhibits NS5-IMPα interaction in a cell context using quantitative bimolecular fluorescence complementation. Finally, we show for the first time that ivermectin can limit infection by the DENV-related West Nile virus at low (µM) concentrations. Since it is FDA approved for parasitic indications, ivermectin merits closer consideration as a broad spectrum antiviral of interest.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Ivermectin/pharmacology , alpha Karyopherins/antagonists & inhibitors , beta Karyopherins/antagonists & inhibitors , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Flavivirus Infections/drug therapy , Kidney/cytology , Protein Binding , Vero Cells , alpha Karyopherins/metabolism , beta Karyopherins/metabolism
13.
Rev Med Virol ; 30(4): e2100, 2020 07.
Article in English | MEDLINE | ID: mdl-32101633

ABSTRACT

Flavivirus infections are a public health threat in the world that requires the development of safe and effective vaccines. Therefore, the understanding of the anti-flavivirus humoral immune response is fundamental to future studies on flavivirus pathogenesis and the design of anti-flavivirus therapeutics. This review aims to provide an overview of the current understanding of the function and involvement of flavivirus proteins in the humoral immune response as well as the ability of the anti-envelope (anti-E) antibodies to interfere (neutralizing antibodies) or not (non-neutralizing antibodies) with viral infection, and how they can, in some circumstances enhance dengue virus infection on Fc gamma receptor (FcγR) bearing cells through a mechanism known as antibody-dependent enhancement (ADE). Thus, the dual role of the antibodies against E protein poses a formidable challenge for vaccine development. Also, we discuss the roles of antibody binding stoichiometry (the concentration, affinity, or epitope recognition) in the neutralization of flaviviruses and the "breathing" of flavivirus virions in the humoral immune response. Finally, the relevance of some specific antibodies in the design and improvement of effective vaccines is addressed.


Subject(s)
Disease Susceptibility/immunology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Flavivirus/immunology , Host-Pathogen Interactions/immunology , Immunity, Humoral/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Flavivirus/drug effects , Flavivirus Infections/drug therapy , Humans , Viral Envelope Proteins/immunology , Viral Vaccines/immunology
14.
Curr Drug Discov Technol ; 17(4): 484-497, 2020.
Article in English | MEDLINE | ID: mdl-31969106

ABSTRACT

Arboviruses are a diverse group of viruses that are among the major causes of emerging infectious diseases. Arboviruses from the genera flavivirus and alphavirus are the most important human arboviruses from a public health perspective. During recent decades, these viruses have been responsible for millions of infections and deaths around the world. Over the past few years, several investigations have been carried out to identify antiviral agents to treat these arbovirus infections. The use of synthetic antiviral compounds is often unsatisfactory since they may raise the risk of viral mutation; they are costly and possess either side effects or toxicity. One attractive strategy is the use of plants as promising sources of novel antiviral compounds that present significant inhibitory effects on these viruses. In this review, we describe advances in the exploitation of compounds and extracts from natural sources that target the vital proteins and enzymes involved in arbovirus replication.


Subject(s)
Alphavirus Infections/drug therapy , Antiviral Agents/pharmacology , Flavivirus Infections/drug therapy , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Alphavirus/drug effects , Alphavirus/genetics , Alphavirus/pathogenicity , Alphavirus Infections/transmission , Alphavirus Infections/virology , Animals , Antiviral Agents/therapeutic use , Disease Reservoirs/virology , Disease Vectors , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Flavivirus/drug effects , Flavivirus/genetics , Flavivirus/pathogenicity , Flavivirus Infections/transmission , Flavivirus Infections/virology , Humans , Mutation , Phytochemicals/therapeutic use , Plant Extracts/therapeutic use , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Virus Replication/drug effects
15.
Virus Res ; 274: 197770, 2019 12.
Article in English | MEDLINE | ID: mdl-31626874

ABSTRACT

Flaviviruses are group of single stranded RNA viruses that cause severe endemic infection and epidemics on a global scale. It presents a significant health impact worldwide and the viruses have the potential to emerge and outbreak in a non-endemic geographical region. Effective vaccines for prophylaxis are only available for several flaviviruses such as Yellow Fever virus, Tick-borne Encephalitis Virus, Dengue Virus and Japanese Encephalitis Virus and there is no antiflaviviral agent being marketed. This review discusses the flavivirus genome, replication cycle, epidemiology, clinical presentation and pathogenesis upon infection. Effective humoral response is critical to confer protective immunity against flaviviruses. Hence, we have also highlighted the immune responses elicited upon infection, various diagnostic facilities available for flaviviral disease and monoclonal antibodies available to date against flavivirus infection.


Subject(s)
Flavivirus Infections/diagnosis , Flavivirus Infections/immunology , Flavivirus/immunology , Flavivirus/isolation & purification , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Biomarkers/blood , Flavivirus/genetics , Flavivirus/physiology , Flavivirus Infections/drug therapy , Flavivirus Infections/pathology , Genome, Viral , Humans , Viral Nonstructural Proteins/blood , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/therapeutic use , Virus Replication
16.
Vet Microbiol ; 235: 295-300, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31383316

ABSTRACT

Tembusu virus (TMUV), a member of the genus flavivirus, primarily causes egg-drop syndrome in ducks and is associated with low disease mortality but high morbidity. The commercially available live vaccines for treating TMUV currently include the main WF100, HB, and FX2010-180P strains, and efficient treatment and/or preventative measures are still urgently needed. Capsid-targeted viral inactivation (CTVI) is a conceptually powerful new antiviral strategy that is based on two proteins from the capsid protein of a virus and a crucial effector molecule. The effector molecule can destroy the viral DNA/RNA or interfere with the proper folding of key viral proteins, while the capsid protein mainly plays a role in viral integration and assembly; the fusion proteins are incorporated into virions during packaging. This study aimed to explore the potential use of this strategy in duck TMUV. Our results revealed that these fusion proteins can be expressed in susceptible BHK21 cells without cytotoxicity and possess excellent Ca2+-dependent nuclease activity, and their expression is also detectable in DF-1 cells. Compared to those in the negative controls (BHK21 and BHK21/pcDNA3.1(+) cells), the numbers of viral RNA copies in TMUV-infected BHK21/Cap-SNase and BHK21/Cap-Linker-SNase cells were reduced by 48 h, and the effect of Cap-Linker-SNase was superior to that of Cap-SNase. As anticipated, these results suggest that these fusion proteins contribute to viral resistance to treatment. Thus, CTVI might be applicable for TMUV inhibition as a novel antiviral therapeutic candidate during viral infection.


Subject(s)
Capsid Proteins/pharmacology , Micrococcal Nuclease/pharmacology , Viral Fusion Proteins/pharmacology , Virus Inactivation/drug effects , Virus Replication/drug effects , Animals , Capsid Proteins/genetics , Cell Line , Ducks , Flavivirus , Flavivirus Infections/drug therapy , Flavivirus Infections/virology , Micrococcal Nuclease/genetics
17.
PLoS Pathog ; 15(8): e1007949, 2019 08.
Article in English | MEDLINE | ID: mdl-31374104

ABSTRACT

Host encounters with viruses lead to an innate immune response that must be rapid and broadly targeted but also tightly regulated to avoid the detrimental effects of unregulated interferon expression. Viral stimulation of host negative regulatory mechanisms is an alternate method of suppressing the host innate immune response. We examined three key mediators of the innate immune response: NF-KB, STAT1 and STAT2 during HCV infection in order to investigate the paradoxical induction of an innate immune response by HCV despite a multitude of mechanisms combating the host response. During infection, we find that all three are repressed only in HCV infected cells but not in uninfected bystander cells, both in vivo in chimeric mouse livers and in cultured Huh7.5 cells after IFNα treatment. We show here that HCV and Flaviviruses suppress the innate immune response by upregulation of PDLIM2, independent of the host interferon response. We show PDLIM2 is an E3 ubiquitin ligase that also acts to stimulate nuclear degradation of STAT2. Interferon dependent relocalization of STAT1/2 to the nucleus leads to PDLIM2 ubiquitination of STAT2 but not STAT1 and the proteasome-dependent degradation of STAT2, predominantly within the nucleus. CRISPR/Cas9 knockout of PDLIM2 results in increased levels of STAT2 following IFNα treatment, retention of STAT2 within the nucleus of HCV infected cells after IFNα stimulation, increased interferon response, and increased resistance to infection by several flaviviruses, indicating that PDLIM2 is a global regulator of the interferon response.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Flavivirus Infections/immunology , Flavivirus/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Immunity, Innate/immunology , LIM Domain Proteins/physiology , STAT2 Transcription Factor/metabolism , Animals , Antiviral Agents/pharmacology , Flavivirus/drug effects , Flavivirus Infections/drug therapy , Flavivirus Infections/virology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Immunity, Innate/drug effects , Interferon-alpha/pharmacology , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NF-kappa B , STAT2 Transcription Factor/genetics , Signal Transduction
18.
Viruses ; 11(6)2019 06 17.
Article in English | MEDLINE | ID: mdl-31212939

ABSTRACT

Chronic viral disease constitutes a major global health problem, with several hundred million people affected and an associated elevated number of deaths. An increasing number of disorders caused by human flaviviruses are related to their capacity to establish a persistent infection. Here we show that Usutu virus (USUV), an emerging zoonotic flavivirus linked to sporadic neurologic disease in humans, can establish a persistent infection in cell culture. Two independent lineages of Vero cells surviving USUV lytic infection were cultured over 82 days (41 cell transfers) without any apparent cytopathology crisis associated. We found elevated titers in the supernatant of these cells, with modest fluctuations during passages but no overall tendency towards increased or decreased infectivity. In addition to full-length genomes, viral RNA isolated from these cells at passage 40 revealed the presence of defective genomes, containing different deletions at the 5' end. These truncated transcripts were all predicted to encode shorter polyprotein products lacking membrane and envelope structural proteins, and most of non-structural protein 1. Treatment with different broad-range antiviral nucleosides revealed that USUV is sensitive to these compounds in the context of a persistent infection, in agreement with previous observations during lytic infections. The exposure of infected cells to prolonged treatment (10 days) with favipiravir and/or ribavirin resulted in the complete clearance of infectivity in the cellular supernatants (decrease of ~5 log10 in virus titers and RNA levels), although modest changes in intracellular viral RNA levels were recorded (<2 log10 decrease). Drug withdrawal after treatment day 10 resulted in a relapse in virus titers. These results encourage the use of persistently-infected cultures as a surrogate system in the identification of improved antivirals against flaviviral chronic disease.


Subject(s)
Flavivirus Infections/virology , Flavivirus/growth & development , Models, Biological , Virus Cultivation/methods , Virus Replication , Amides/administration & dosage , Amides/pharmacology , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Chlorocebus aethiops , Flavivirus Infections/drug therapy , Pyrazines/administration & dosage , Pyrazines/pharmacology , RNA, Viral/genetics , Ribavirin/administration & dosage , Ribavirin/pharmacology , Sequence Analysis, DNA , Sequence Deletion , Serial Passage , Treatment Outcome , Vero Cells , Viral Load , Viral Proteins/genetics
19.
Nat Microbiol ; 4(6): 985-995, 2019 06.
Article in English | MEDLINE | ID: mdl-30833725

ABSTRACT

West Nile virus (WNV) is an emerging mosquito-borne flavivirus, related to dengue virus and Zika virus. To gain insight into host pathways involved in WNV infection, we performed a systematic affinity-tag purification mass spectrometry (APMS) study to identify 259 WNV-interacting human proteins. RNA interference screening revealed 26 genes that both interact with WNV proteins and influence WNV infection. We found that WNV, dengue and Zika virus capsids interact with a conserved subset of proteins that impact infection. These include the exon-junction complex (EJC) recycling factor PYM1, which is antiviral against all three viruses. The EJC has roles in nonsense-mediated decay (NMD), and we found that both the EJC and NMD are antiviral and the EJC protein RBM8A directly binds WNV RNA. To counteract this, flavivirus infection inhibits NMD and the capsid-PYM1 interaction interferes with EJC protein function and localization. Depletion of PYM1 attenuates RBM8A binding to viral RNA, suggesting that WNV sequesters PYM1 to protect viral RNA from decay. Together, these data suggest a complex interplay between the virus and host in regulating NMD and the EJC.


Subject(s)
Antiviral Agents/pharmacology , Flavivirus Infections/drug therapy , Viral Proteins/genetics , West Nile virus/drug effects , West Nile virus/genetics , Capsid Proteins , Carrier Proteins , Codon, Nonsense , Dengue Virus/genetics , Exons , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Protein Interaction Maps , RNA Interference , RNA, Viral , RNA-Binding Proteins , Viral Proteins/physiology , West Nile virus/pathogenicity , Zika Virus/genetics
20.
ACS Infect Dis ; 5(3): 460-472, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30608640

ABSTRACT

Vaccines and antivirals to combat dengue, Zika, and other flavivirus pathogens present a major, unmet medical need. Vaccine development has been severely challenged by the antigenic diversity of these viruses and the propensity of non-neutralizing, cross-reactive antibodies to facilitate cellular infection and increase disease severity. As an alternative, direct-acting antivirals targeting the flavivirus envelope protein, E, have the potential to act via an analogous mode of action without the risk of antibody-dependent enhancement of infection and disease. We previously discovered that structurally diverse small molecule inhibitors of the dengue virus E protein exhibit varying levels of antiviral activity against other flaviviruses in cell culture. Here, we demonstrate that the broad-spectrum activity of several cyanohydrazones against dengue, Zika, and Japanese encephalitis viruses is due to specific inhibition of E-mediated membrane fusion during viral entry and provide proof of concept for pharmacological inhibition of E as an antiviral strategy in vivo.


Subject(s)
Antiviral Agents/administration & dosage , Flavivirus Infections/drug therapy , Flavivirus/drug effects , Small Molecule Libraries/administration & dosage , Viral Envelope Proteins/metabolism , Animals , Antiviral Agents/chemistry , Female , Flavivirus/physiology , Flavivirus Infections/virology , Humans , Male , Mice , Mice, Inbred C57BL , Small Molecule Libraries/chemistry , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/genetics , Virus Internalization/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...