Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
1.
Int J Biol Macromol ; 267(Pt 1): 131453, 2024 May.
Article in English | MEDLINE | ID: mdl-38588842

ABSTRACT

Enterovirus 71 (EV71) causes hand-foot-and-mouth disease (HFMD), neurological complications, and even fatalities in infants. Clinically, the increase of extracellular vesicles (EVs) in EV71 patients' serum was highly associated with the severity of HFMD. EV71 boosts EVs biogenesis in an endosomal sorting complex required for transport (ESCRT)-dependent manner to facilitate viral replication. Yet, the impact of EVs-derived from ESCRT-independent pathway on EV71 replication and pathogenesis is highly concerned. Here, we assessed the effects of EV71-induced EVs from ESCRT-independent pathway on viral replication and pathogenesis by GW4869, a neutral sphingomyelinase inhibitor. Detailly, in EV71-infected mice, blockade of the biogenesis of tissue-derived EVs in the presence of GW4869 restored body weight loss, attenuated clinical scores, and improved survival rates. Furthermore, GW4869 dampens EVs biogenesis to reduce viral load and pathogenesis in multiple tissues of EV71-infected mice. Consistently, GW4869 treatment in a human intestinal epithelial HT29 cells decreased the biogenesis of EVs, in which the progeny EV71 particle was cloaked, leading to the reduction of viral infection and replication. Collectively, GW4869 inhibits EV71-induced EVs in an ESCRT-independent pathway and ultimately suppresses EV71 replication and pathogenesis. Our study provides a novel strategy for the development of therapeutic agents in the treatment for EV71-associated HFMD.


Subject(s)
Aniline Compounds , Endosomal Sorting Complexes Required for Transport , Enterovirus A, Human , Extracellular Vesicles , Virus Replication , Animals , Virus Replication/drug effects , Enterovirus A, Human/drug effects , Enterovirus A, Human/physiology , Mice , Extracellular Vesicles/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Benzylidene Compounds/pharmacology , Enterovirus Infections/virology , Enterovirus Infections/drug therapy , Enterovirus Infections/metabolism , Viral Load/drug effects , Female
2.
Virol Sin ; 39(2): 301-308, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38452856

ABSTRACT

Hand, foot, and mouth disease (HFMD) is a common pediatric illness mainly caused by enteroviruses, which are important human pathogens. Currently, there are no available antiviral agents for the therapy of enterovirus infection. In this study, an excellent high-content antiviral screening system utilizing the EV-A71-eGFP reporter virus was developed. Using this screening system, we screened a drug library containing 1042 natural compounds to identify potential EV-A71 inhibitors. Fangchinoline (FAN), a bis-benzylisoquinoline alkaloid, exhibits potential inhibitory effects against various enteroviruses that cause HFMD, such as EV-A71, CV-A10, CV-B3 and CV-A16. Further investigations revealed that FAN targets the early stage of the enterovirus life cycle. Through the selection of FAN-resistant EV-A71 viruses, we demonstrated that the VP1 protein could be a potential target of FAN, as two mutations in VP1 (E145G and V258I) resulted in viral resistance to FAN. Our research suggests that FAN is an efficient inhibitor of EV-A71 and has the potential to be a broad-spectrum antiviral drug against human enteroviruses.


Subject(s)
Antiviral Agents , Benzylisoquinolines , Drug Resistance, Viral , Antiviral Agents/pharmacology , Humans , Benzylisoquinolines/pharmacology , Drug Resistance, Viral/genetics , Virus Replication/drug effects , Enterovirus A, Human/drug effects , Enterovirus A, Human/genetics , Drug Evaluation, Preclinical , Genes, Reporter , High-Throughput Screening Assays , Capsid Proteins/genetics , Capsid Proteins/antagonists & inhibitors , Enterovirus/drug effects , Enterovirus/genetics , Cell Line , Green Fluorescent Proteins/genetics
3.
Bioorg Chem ; 122: 105683, 2022 05.
Article in English | MEDLINE | ID: mdl-35278779

ABSTRACT

Enterovirus A71 (EV-A71), one of the major pathogens that causes hand, foot and mouth disease (HFMD), has seriously threatened the health and safety of young children. In this study, aminothiazole derivatives were synthesized and screened against EV-A71 in Rhabdomyosarcoma (RD) cells. The best compound (12s), with a biphenyl group, showed activity against EV-A71 (EC50: 0.27 µM) but also against a series of different human enteroviruses without significant cytotoxicity (CC50 > 56.2 µM). Mechanistic studies including time-of-drug-addition assays, viral entry assays and microscale thermophoresis (MST) experiments, showed that 12s binds to EV-A71 capsid and blocks the binding between the viral protein VP1 and the relevant human scavenger receptor class B member 2 (hSCARB2).


Subject(s)
Capsid Proteins , Enterovirus A, Human , Thiazoles , Capsid Proteins/antagonists & inhibitors , Enterovirus A, Human/drug effects , Enterovirus Infections/drug therapy , Humans , Thiazoles/pharmacology , Virus Internalization
4.
Biochem Pharmacol ; 194: 114820, 2021 12.
Article in English | MEDLINE | ID: mdl-34748818

ABSTRACT

Human enterovirus A71 (EV-A71) is a major etiological agent of hand-foot-and-mouth disease (HFMD) and there is presently no internationally approved antiviral against EV-A71. In this study, it is disclosed that 14S-(2'-chloro-4'-nitrophenoxy)-8R/S,17-epoxy andrographolide (2) was discovered to be an effective inhibitor against EV-A71 infection showing significant reduction of viral titre. In addition to EV-A71, compound 2 exerts broad-spectrum antiviral effects against other enteroviruses. It is revealed that compound 2 inhibits the post-entry stages of EV-A71 viral replication cycle and significantly reduces viral protein expression of structural proteins such as VP0 and VP2 via inhibiting EV-A71 RNA replication. Moreover, the inhibitory property of compound 2 is specific to viral RNA replication. Furthermore, compound 2 is more likely to target a host factor in EV-A71 RNA replication. As a result, introduction of epoxide at positions 8 and 17 of andrographolide is effective for anti-EV-A71 infection and is a potential anti-EV-A71 strategy. Further work to discover more potent andrographolide derivatives and elucidate comprehensive SAR is under way.


Subject(s)
Diterpenes/pharmacology , Drug Discovery/methods , Enterovirus A, Human/drug effects , Enterovirus Infections , Virus Replication/drug effects , Animals , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Diterpenes/chemistry , Diterpenes/therapeutic use , Dose-Response Relationship, Drug , Enterovirus A, Human/physiology , Enterovirus Infections/drug therapy , Enterovirus Infections/metabolism , Humans , Vero Cells , Virus Replication/physiology
5.
Molecules ; 26(21)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34770987

ABSTRACT

Enterovirus 71 (EV-A71) is one of the predominant etiological agents of hand, foot and mouth disease (HMFD), which can cause severe central nervous system infections in young children. There is no clinically approved vaccine or antiviral agent against HFMD. The SP40 peptide, derived from the VP1 capsid of EV-A71, was reported to be a promising antiviral peptide that targeted the host receptor(s) involved in viral attachment or entry. So far, the mechanism of action of SP40 peptide is unknown. In this study, interactions between ten reported cell receptors of EV-A71 and the antiviral SP40 peptide were evaluated through molecular docking simulations, followed by in vitro receptor blocking with specific antibodies. The preferable binding region of each receptor to SP40 was predicted by global docking using HPEPDOCK and the cell receptor-SP40 peptide complexes were refined using FlexPepDock. Local molecular docking using GOLD (Genetic Optimization for Ligand Docking) showed that the SP40 peptide had the highest binding score to nucleolin followed by annexin A2, SCARB2 and human tryptophanyl-tRNA synthetase. The average GoldScore for 5 top-scoring models of human cyclophilin, fibronectin, human galectin, DC-SIGN and vimentin were almost similar. Analysis of the nucleolin-SP40 peptide complex showed that SP40 peptide binds to the RNA binding domains (RBDs) of nucleolin. Furthermore, receptor blocking by specific monoclonal antibody was performed for seven cell receptors of EV-A71 and the results showed that the blocking of nucleolin by anti-nucleolin alone conferred a 93% reduction in viral infectivity. Maximum viral inhibition (99.5%) occurred when SCARB2 was concurrently blocked with anti-SCARB2 and the SP40 peptide. This is the first report to reveal the mechanism of action of SP40 peptide in silico through molecular docking analysis. This study provides information on the possible binding site of SP40 peptide to EV-A71 cellular receptors. Such information could be useful to further validate the interaction of the SP40 peptide with nucleolin by site-directed mutagenesis of the nucleolin binding site.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Molecular Docking Simulation , Peptides/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Enterovirus A, Human/metabolism , Humans , Peptides/chemical synthesis , Peptides/chemistry , Receptors, Cell Surface/metabolism , Software
6.
Microbiol Spectr ; 9(3): e0102521, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34787443

ABSTRACT

The fatal pathogen enterovirus 71 (EV71) is a major cause of hand-foot-and-mouth disease (HFMD), which leads to serious neurological syndromes. While there are no effective clinical agents available for EV71 treatment thus far, EV71 3C protease (3Cpro), a cysteine protease encoded by the virus, has become a promising drug target for discovery of antiviral drugs, given that it plays a crucial role in virus proliferation and interferes with host cell function. Here, we report two inhibitors of EV71 3Cpro, FOPMC and FIOMC, that were developed from previously reported cyanohydrin derivative (R)-1 by replacing the acyl cyanohydrin group with 4-iminooxazolidin-2-one. FOPMC and FIOMC have potent antiviral activity and dramatically improved metabolic stability. These two inhibitors demonstrated broad anti-EV effects on various cell lines and five epidemic viral strains. We further illuminated the binding models between 3Cpro and FOPMC/FIOMC through molecular docking and molecular dynamics simulations. The substitution of an acyl cyanohydrin group with 4-iminooxazolidin-2-one does make FOPMC and FIOMC potent anti-EV71 drug candidates as universal nonclassical bioisosteres with a cyanohydrin moiety. IMPORTANCE EV71 is one of the most epidemic agents of HFMD. Thus far, there are no antiviral drugs available for clinical usage. The conserved EV71 3Cpro plays pivotal roles in virus proliferation and defense host immunity, as well as having no homology in host cells, making it a most promising antiviral target. In this work, we identified that propyl- and isopropyl-substituted 4-iminooxazolidin-2-one moieties (FOPMC and FIOMC) effectively inhibited five epidemic viral strains in rhabdomyosarcoma (RD), HEK-293T, and VeroE6 cell lines. The inhibition mechanism was also illustrated with molecular docking and molecular dynamics (MD) simulations. The successful replacement of the labile cyanohydrin greatly improved the stability and pharmacokinetic properties of (R)-1, making 4-iminooxazolidin-2-one a nonclassical bioisosteric moiety of cyanohydrin. This discovery addressed a critical issue of the primitive structural scaffold of these promising anti-EV71 inhibitors and could lead to their development as broad-spectrum anti-EV agents.


Subject(s)
3C Viral Proteases , Antiviral Agents , Enterovirus A, Human , Virus Replication , Animals , Humans , 3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line, Tumor , Chlorocebus aethiops , Enterovirus A, Human/drug effects , Enterovirus A, Human/growth & development , Hand, Foot and Mouth Disease/drug therapy , Hand, Foot and Mouth Disease/prevention & control , Hand, Foot and Mouth Disease/virology , HEK293 Cells , Molecular Docking Simulation , Molecular Dynamics Simulation , Nitriles/chemistry , Nitriles/pharmacology , Vero Cells , Virus Replication/drug effects
7.
Life Sci ; 287: 120097, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34715144

ABSTRACT

AIMS: Enterovirus A71 (EV-A71) is an etiological agent of hand foot and mouth disease (HFMD) and has the potential to cause severe neurological infections in children. L-SP40 peptide was previously known to inhibit EV-A71 by prophylactic action. This study aimed to identify the mechanism of inhibition in Rhabdomyosarcoma (RD) cells and in vivo therapeutic potential of L-SP40 peptide in a murine model. MAIN METHODS: A pull-down assay was performed to identify the binding partner of the L-SP40 peptide. Co-immunoprecipitation and co-localization assays with the L-SP40 peptide were employed to confirm the receptor partner in RD cells. The outcomes were validated using receptor knockdown and antibody blocking assays. The L-SP40 peptide was further evaluated for the protection of neonatal mice against lethal challenge by mouse-adapted EV-A71. KEY FINDINGS: The L-SP40 peptide was found to interact and co-localize with nucleolin, the key attachment receptor of Enteroviruses A species, as demonstrated in the pull-down, co-immunoprecipitation and co-localization assays. Knockdown of nucleolin from RD cells led to a significant reduction of 3.5 logs of viral titer of EV-A71. The L-SP40 peptide demonstrated 80% protection of neonatal mice against lethal challenge by the mouse-adapted virus with a drastic reduction in the viral loads in the blood (~4.5 logs), skeletal muscles (1.5 logs) and brain stem (1.5 logs). SIGNIFICANCE: L-SP40 peptide prevented severe hind limb paralysis and death in suckling mice and could serve as a potential broad-spectrum antiviral candidate to be further evaluated for safety and potency in future clinical trials against EV-A71.


Subject(s)
Enterovirus A, Human/drug effects , Enterovirus A, Human/metabolism , Enterovirus Infections/drug therapy , Enterovirus Infections/metabolism , Peptide Fragments/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Animals, Newborn , Mice , Mice, Inbred ICR , Peptide Fragments/administration & dosage , Protein Binding/physiology , Treatment Outcome , Nucleolin
8.
Trop Biomed ; 38(3): 239-247, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34362866

ABSTRACT

Hand, foot and mouth disease (HFMD) is a highly contagious viral disease that predominantly affects children younger than 5 years old. HFMD is primarily caused by enterovirus A71 (EVA71) and coxsackievirus A16 (CV-A16). However, coxsackievirus A10 (CV-A10) and coxsackievirus A6 (CV-A6) are being increasingly reported as the predominant causative of HFMD outbreaks worldwide since the past decade. To date, there are still no licensed multivalent vaccines or antiviral drugs targeting enteroviruses that cause HFMD, despite HFMD outbreaks are still being frequently reported, especially in Asia-Pacific countries. The high rate of transmission, morbidity and potential neurological complications of HFMD is indeed making the development of broad-spectrum antiviral drugs/agents against these enteroviruses a compelling need. In this study, we have investigated the in vitro antiviral effect of 4 Ganoderma neo-japonicum Imazeki (GNJI) crude extracts (S1-S4) against EV-A71, CV-A16, CV-A10 and CV-A6. GNJI is a medicinal mushroom that can be found growing saprophytically on decaying bamboo clumps in Malaysian forests. The antiviral effects of this medicinal mushroom were determined using cytopathic inhibition and virus titration assays. The S2 (1.25 mg/ml) hot aqueous extract demonstrated the highest broad-spectrum antiviral activity against all tested enteroviruses in human primary oral fibroblast cells. Replication of EV-A71, CV-A16 and CVA10 were effectively inhibited at 2 hours post-infection (hpi) to 72 hpi, except for CV-A6 which was only at 2 hpi. S2 also has virucidal activity against EV-A71. Polysaccharides isolated and purified from crude hot aqueous extract demonstrated similar antiviral activity as S2, suggesting that polysaccharides could be one of the active compounds responsible for the antiviral activity shown by S2. To our knowledge, this study demonstrates for the first time the ability of GNJI to inhibit enterovirus infection and replication. Thus, GNJI is potential to be further developed as an antiviral agent against enteroviruses that caused HFMD.


Subject(s)
Antiviral Agents , Biological Products/pharmacology , Enterovirus A, Human , Ganoderma , Antiviral Agents/pharmacology , Cells, Cultured , China , Enterovirus A, Human/drug effects , Enterovirus Infections , Fibroblasts/virology , Ganoderma/chemistry , Hand, Foot and Mouth Disease , Humans
9.
Virol J ; 18(1): 164, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34384436

ABSTRACT

BACKGROUND: Enterovirus 71 (EV-A71) is a highly infectious pathogen associated with hand, foot and mouth disease, herpangina, and various neurological complications, so it is important for the early detection and treatment of EV-A71. An aptamer is a nucleotide sequence that screened in vitro by the technology named systematic evolution of ligands by exponential enrichment technology (SELEX). Similar to antibodies, aptamers can bind to the targets with high specificity and affinity. Besides, emerging aptamers have many advantages comparing with antibodies, such as ease of synthesis and modification, having a wide variety of target materials, low manufacturing cost and easy flexibility in amending. Therefore, aptamers are promising in virus detection and anti-virus therapy. METHODS: Aptamers were selected by SELEX. Specificity, affinity and second structure were used to characterize the selected aptamers. Chemiluminescence was adopted to build an aptamer-based detection method for EV-A71. Cytopathogenic effects trial, the level of intracellular EV-A71 RNA and protein expression were used to evaluate the antiviral effect of the selected aptamers. RESULTS: Three DNA aptamers with high specificity and affinity for EV-A71structual protein VP1 were screened out. A rapid chemiluminutesescence aptamer biosensor for EV-A71 detection was designed out. The selected aptamers could inhibit the RNA replication and protein expression of EV-A71 in RD cells and ameliorate the cytopathogenic effects. CONCLUSIONS: The aptamers against EV-A71 have the potentiality to be applied as attractive candidates used for EV-A71 detection and treatment in the future.


Subject(s)
Aptamers, Nucleotide , Enterovirus A, Human , Aptamers, Nucleotide/pharmacology , Capsid Proteins , Enterovirus A, Human/drug effects , Enterovirus Infections , Humans , RNA
10.
Antiviral Res ; 194: 105164, 2021 10.
Article in English | MEDLINE | ID: mdl-34411654

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an acute infectious disease caused by a novel phlebovirus (SFTSV), characterized by fever, thrombocytopenia and leukocytopenia which lead to multiple organ failure with high mortality in severe cases. The SFTSV has spread rapidly in recent years and posed a serious threat to public health in endemic areas. However, specific antiviral therapeutics for SFTSV infection are rare. In this study, we demonstrated that two peptides, SGc1 and SGc8, derived from a hydrophobic region of the SFTSV glycoprotein Gc, could potently inhibit SFTSV replication in a dose-dependent manner without apparent cytotoxicity in various cell lines and with low immunogenicity and good stability. The IC50 (50% inhibition concentration) values for both peptides to inhibit 2 MOI of SFTSV infection were below 10 µM in L02, Vero and BHK21 cells. Mechanistically, SGc1 and SGc8 mainly inhibited viral entry at the early stage of the viral infection. Inhibition of SFTSV replication was specific by both peptides because no inhibitory effect was shown against other viruses including Zika virus and Enterovirus A71. Taken together, our results suggested that viral glycoprotein-derived SGc1 and SGc8 peptides have antiviral potential and warrant further assessment as an SFTSV-specific therapeutic.


Subject(s)
Antiviral Agents/pharmacology , Glycoproteins/pharmacology , Peptides/pharmacology , Phlebovirus/chemistry , Phlebovirus/drug effects , Viral Nonstructural Proteins/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Enterovirus A, Human/drug effects , Female , Glycoproteins/chemistry , Inhibitory Concentration 50 , Mice , Peptides/chemistry , Phlebovirus/genetics , Severe Fever with Thrombocytopenia Syndrome/drug therapy , Vero Cells , Virus Internalization/drug effects , Virus Replication/drug effects , Zika Virus/drug effects
11.
J Med Chem ; 64(14): 10027-10046, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34229438

ABSTRACT

We have recently described a new generation of potent human immunodeficiency virus (HIV) and EV-A71 entry inhibitors. The prototypes contain three or four tryptophan (Trp) residues bearing an isophthalic acid moiety at the C2 position of each side-chain indole ring. This work is now extended by both shifting the position of the isophthalic acid to C7 and synthesizing doubly arylated C2/C7 derivatives. The most potent derivative (50% effective concentration (EC50) HIV-1, 6 nM; EC50 EV-A71, 40 nM), 33 (AL-518), is a C2/C7 doubly arylated tetrapodal compound. Its superior anti-HIV potency with respect to the previous C2-arylated prototype is in consonance with its higher affinity for the viral gp120. 33 (AL-518) showed comparable antiviral activities against X4 and R5 HIV-1 strains and seems to interact with the tip and base of the gp120 V3 loop. Taken together, these findings support the interest in 33 (AL-518) as a useful new prototype for anti-HIV/EV71 drug development.


Subject(s)
Anti-HIV Agents/pharmacology , Enterovirus A, Human/drug effects , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Indoles/pharmacology , Tryptophan/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Dose-Response Relationship, Drug , HIV Fusion Inhibitors/chemical synthesis , HIV Fusion Inhibitors/chemistry , Indoles/chemistry , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Tryptophan/chemical synthesis , Tryptophan/chemistry
12.
J Med Chem ; 64(12): 8755-8774, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34085827

ABSTRACT

The enterovirus genus of the picornavirus family contains many important human pathogens. EV-D68 primarily infects children, and the disease manifestations range from respiratory illnesses to neurological complications such as acute flaccid myelitis (AFM). EV-A71 is a major pathogen for the hand, foot, and mouth disease (HFMD) in children and can also lead to AFM and death in severe cases. CVB3 infection can cause cardiac arrhythmias, acute heart failure, as well as type 1 diabetes. There is currently no FDA-approved antiviral for any of these enteroviruses. In this study, we report our discovery and development of pyrazolopyridine-containing small molecules with potent and broad-spectrum antiviral activity against multiple strains of EV-D68, EV-A71, and CVB3. Serial viral passage experiments, coupled with reverse genetics and thermal shift binding assays, suggested that these molecules target the viral protein 2C. Overall, the pyrazolopyridine inhibitors represent a promising class of candidates for the urgently needed nonpolio enterovirus antivirals.


Subject(s)
Antiviral Agents/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Antiviral Agents/chemical synthesis , Carrier Proteins , Cell Line, Tumor , Enterovirus A, Human/drug effects , Enterovirus B, Human/drug effects , Enterovirus D, Human/drug effects , Humans , Microbial Sensitivity Tests , Molecular Structure , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Structure-Activity Relationship , Viral Nonstructural Proteins
13.
Antiviral Res ; 192: 105102, 2021 08.
Article in English | MEDLINE | ID: mdl-34082057

ABSTRACT

As one of the principal etiological agents of hand, foot, and mouth disease (HFMD), enterovirus 71 (EV71) is associated with severe neurological complications or fatal diseases, while without effective medications thus far. Here we applied dually activated Michael acceptor to develop a series of reversible covalent compounds for EV71 3C protease (3Cpro), a promising antiviral drug target that plays an essential role during viral replication by cleaving the precursor polyprotein, inhibiting host protein synthesis, and evading innate immunity. Among them, cyanoacrylate and Boc-protected cyanoarylamide derivatives (SLQ-4 and SLQ-5) showed effective antiviral activity against EV71. The two inhibitors exhibited broad antiviral effects, acting on RD, 293T, and Vero cell lines, as well as on EV71 A, B, C, CVA16, and CVB3 viral strains. We further determined the binding pockets between the two inhibitors and 3Cpro based on docking studies. These results, together with our previous studies, provide evidence to elucidate the mechanism of action of these two reversible covalent inhibitors and contribute to the development of clinically effective medicines to treat EV71 infections.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Protease Inhibitors/pharmacology , 3C Viral Proteases/chemistry , Acrylamides/chemistry , Acrylamides/pharmacology , Animals , Antiviral Agents/chemistry , Cell Line , Cell Survival/drug effects , Cyanoacrylates/chemistry , Cyanoacrylates/pharmacology , Enterovirus/classification , Enterovirus/drug effects , Enterovirus Infections/virology , Humans , Molecular Docking Simulation , Protease Inhibitors/chemistry , Virus Replication/drug effects
14.
Antiviral Res ; 195: 105091, 2021 11.
Article in English | MEDLINE | ID: mdl-34044060

ABSTRACT

Enterovirus A71 (EV-A71) is one of the main causative agents of hand-foot-mouth disease (HFMD) and causes serious neurological complications. However, no effective therapy is currently available for treating these infections. Therefore, effective drugs to prevent and treat EV-A71 infections are urgently needed. Here, we demonstrated that treatment with Licochalcone A (LCA) significantly inhibited EV-A71 replication in a dose-dependent manner, with an EC50 of 9.30 µM in RD cells and 5.73 µM in Vero cells. The preliminary results on the inhibition mechanism showed that LCA exerted antiviral effects by interfering with the early step of viral replication. We further demonstrated that LCA showed potent antiviral activity against many enteroviruses, including EV-A71 (strain C4), EV-A71 (strain H), and coxsackievirus A16 (CV-A16). Furthermore, LCA could effectively prevent the clinical symptoms and death of virus infected mice and decreased viral load in EV-A71-infected mice. Taken together, our studies showed for the first time, that LCA is a promising EV-A71 inhibitor and provide important information for the clinical development of LCA as a potential new anti-EV-A71 agent.


Subject(s)
Antiviral Agents/pharmacology , Chalcones/pharmacology , Enterovirus A, Human/drug effects , Enterovirus Infections/drug therapy , Animals , Animals, Newborn , Cell Line, Tumor , Chlorocebus aethiops , Enterovirus A, Human/growth & development , Hand, Foot and Mouth Disease/drug therapy , Hand, Foot and Mouth Disease/virology , Humans , Mice , Vero Cells , Viral Load/drug effects , Virus Replication
15.
Front Immunol ; 12: 665197, 2021.
Article in English | MEDLINE | ID: mdl-34054834

ABSTRACT

Coxsackievirus A6 (CVA6) is recognized as a major enterovirus type that can cause severe hand, foot, and mouth disease and spread widely among children. Vaccines and antiviral drugs may be developed more effectively based on a stable and easy-to-operate CVA6 mouse infection model. In this study, a wild CVA6-W strain was sub-cultured in newborn mice of different ages (in days), for adaptation. Therefore, a CVA6-A mouse-adapted strain capable of stably infecting the mice was generated, and a fatal model was built. As the result indicated, CVA6-A could infect the 10-day-old mice to generate higher levels of IFN-γ, IL-6, and IL-10. The mice infected with CVA6-A were treated with IFN-α1b at a higher dose, with complete protection. Based on this strain, an animal model with active immunization was built to evaluate antiviral protection by active immunization. The three-day-old mice were pre-immunized with inactivated CVA6 thereby generating IgM and IgG antibodies within 7 days that enabled complete protection of the pre-immunized mice following the CVA6 virus challenge. There were eight mutations in the genome of CVA6-A than in that of CVA6-W, possibly attributed to the virulence of CVA6 in mice. Briefly, the CVA6 infection model of the 10-day-old mice built herein, may serve as an applicable preclinical evaluation model for CVA6 antiviral drugs and vaccine study.


Subject(s)
Antibodies, Viral/therapeutic use , Antiviral Agents/therapeutic use , Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/immunology , Hand, Foot and Mouth Disease/prevention & control , Viral Vaccines/immunology , Animals , Animals, Newborn , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Enterovirus A, Human/drug effects , Enterovirus A, Human/pathogenicity , Hand, Foot and Mouth Disease/drug therapy , Hand, Foot and Mouth Disease/virology , Interferon-gamma/blood , Interferon-gamma/pharmacology , Interleukin-10/blood , Interleukin-10/pharmacology , Interleukin-6/blood , Interleukin-6/pharmacology , Male , Mice , Mice, Inbred BALB C , Vaccination , Vaccines, Inactivated/immunology , Viral Load/drug effects
16.
Eur J Pharmacol ; 906: 174204, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34051220

ABSTRACT

Enterovirus A71 (EV-A71) is one of the main causative agents of hand, foot and mouth disease which seriously threatens young children's health and lives. However, there is no effective therapy currently available for treating these infections. Therefore, effective drugs to prevent and treat EV-A71 infections are urgently needed. Here, we identified Mulberroside C potently against the proliferation of EV-A71. The in-vitro anti-EV-A71 activity of Mulberroside C was assessed by cytopathic effect inhibition and viral plaque reduction assays, and the results showed that Mulberroside C significantly inhibited EV-A71 infection. The downstream assays affirmed that Mulberroside C inhibited viral protein and RNA synthesis. Furthermore, Mulberroside C effectively reduced clinical symptoms in EV-A71 infected mice and reduced mortality at higher concentrations. The mechanism study indicated that Mulberroside C bound to the hydrophobic pocket of viral capsid protein VP1, thereby preventing viral uncoating and genome release. Taken together, our study indicated that Mulberroside C could be a promising EV-A71 inhibitor and worth extensive preclinical investigation as a lead compound.


Subject(s)
Antiviral Agents/pharmacology , Benzopyrans/pharmacology , Enterovirus A, Human/drug effects , Hand, Foot and Mouth Disease/drug therapy , Animals , Animals, Newborn , Antiviral Agents/therapeutic use , Benzopyrans/therapeutic use , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Disease Models, Animal , Enterovirus A, Human/metabolism , Hand, Foot and Mouth Disease/virology , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Morus/chemistry , Specific Pathogen-Free Organisms , Vero Cells , Virus Replication/drug effects
17.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33789997

ABSTRACT

Enteroviruses belong to the genus Enterovirus of the family Picornaviridae and include four human enterovirus groups (EV-A to -D): the epidemic of enteroviruses such as human enterovirus A71 (EV-A71) and coxsackievirus A16 (CVA16) is a threat to global public health. Enteroviral protein 2C is the most conserved nonstructural protein among all enteroviruses and possesses RNA helicase activity that plays pivotal roles during enteroviral life cycles, which makes 2C an attractive target for developing antienterovirus drugs. In this study, we designed a peptide, named 2CL, based on the structure of EV-A71 2C. This peptide effectively impaired the oligomerization of EV-A71 2C protein and inhibited the RNA helicase activities of 2C proteins encoded by EV-A71 and CVA16, both of which belong to EV-A, and showed potent antiviral efficacy against EV-A71 and CVA16 in cells. Moreover, the 2CL treatment elicited a strong in vivo protective efficacy against lethal EV-A71 challenge. In addition, the antiviral strategy of targeting the 2C helicase activity can be applied to inhibit the replication of EV-B. Either 2CL or B-2CL, the peptide redesigned based on the 2CL-corresponding sequence of EV-Bs, could exert effective antiviral activity against two important EV-Bs, coxsackievirus B3 and echovirus 11. Together, our findings demonstrated that targeting the helicase activity of 2C with a rationally designed peptide is an efficient antiviral strategy against enteroviruses, and 2CL and B-2CL show promising clinical potential to be further developed as broad-spectrum antienterovirus drugs.IMPORTANCE Enteroviruses are a large group of positive-sense single-stranded RNA viruses and include numerous human pathogens, such as enterovirus A71 (EV-A71), coxsackieviruses, and echoviruses. However, no approved EV antiviral drugs are available. Enteroviral 2C is the most conserved nonstructural protein among all enteroviruses and contains the RNA helicase activity critical for the viral life cycle. Herein, according to the structure of EV-A71 2C, we designed a peptide that effectively inhibited the RNA helicase activities of EV-A71- and coxsackievirus A16 (CVA16)-encoded 2C proteins. Moreover, this peptide exerted potent antiviral effects against EV-A71 and CVA16 in cells and elicited therapeutic efficacy against lethal EV-A71 challenge in vivo Furthermore, we demonstrate that the strategy of targeting the 2C helicase activity can be used for other relevant enteroviruses, including coxsackievirus B3 and echovirus 11. In summary, our findings provide compelling evidence that the designed peptides targeting the helicase activity of 2C could be broad-spectrum antivirals for enteroviruses.


Subject(s)
Antiviral Agents/pharmacology , Carrier Proteins/antagonists & inhibitors , Enterovirus A, Human/drug effects , Enterovirus Infections/drug therapy , Peptides/pharmacology , RNA Helicases/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Drug Design , Enterovirus A, Human/chemistry , Enterovirus A, Human/physiology , Enterovirus B, Human/drug effects , Enterovirus B, Human/physiology , Enterovirus Infections/virology , Humans , Mice , Mice, Inbred ICR , Peptides/chemistry , Peptides/therapeutic use , RNA Helicases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
18.
Viruses ; 13(5)2021 04 27.
Article in English | MEDLINE | ID: mdl-33925713

ABSTRACT

Viral infections are one of the leading causes in human mortality and disease. Broad-spectrum antiviral drugs are a powerful weapon against new and re-emerging viruses. However, viral resistance to existing broad-spectrum antivirals remains a challenge, which demands development of new broad-spectrum therapeutics. In this report, we showed that fludarabine, a fluorinated purine analogue, effectively inhibited infection of RNA viruses, including Zika virus, Severe fever with thrombocytopenia syndrome virus, and Enterovirus A71, with all IC50 values below 1 µM in Vero, BHK21, U251 MG, and HMC3 cells. We observed that fludarabine has shown cytotoxicity to these cells only at high doses indicating it could be safe for future clinical use if approved. In conclusion, this study suggests that fludarabine could be developed as a potential broad-spectrum anti-RNA virus therapeutic agent.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Phlebovirus/drug effects , Vidarabine/analogs & derivatives , Zika Virus/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Cell Survival , Cells, Cultured , Humans , RNA Viruses/drug effects , Vidarabine/chemistry , Vidarabine/pharmacology , Virus Replication/drug effects
19.
Viruses ; 13(2)2021 02 16.
Article in English | MEDLINE | ID: mdl-33669264

ABSTRACT

Honeysuckle (Lonicera japonica Thunb) is a traditional Chinese medicine (TCM) with an antipathogenic activity. MicroRNAs (miRNAs) are small non-coding RNA molecules that are ubiquitously expressed in cells. Endogenous miRNA may function as an innate response to block pathogen invasion. The miRNA expression profiles of both mice and humans after the ingestion of honeysuckle were obtained. Fifteen overexpressed miRNAs overlapped and were predicted to be capable of targeting three viruses: dengue virus (DENV), enterovirus 71 (EV71) and SARS-CoV-2. Among them, let-7a was examined to be capable of targeting the EV71 RNA genome by reporter assay and Western blotting. Moreover, honeysuckle-induced let-7a suppression of EV71 RNA and protein expression as well as viral replication were investigated both in vitro and in vivo. We demonstrated that let-7a targeted EV71 at the predicted sequences using luciferase reporter plasmids as well as two infectious replicons (pMP4-y-5 and pTOPO-4643). The suppression of EV71 replication and viral load was demonstrated in two cell lines by luciferase activity, RT-PCR, real-time PCR, Western blotting and plaque assay. Furthermore, EV71-infected suckling mice fed honeysuckle extract or inoculated with let-7a showed decreased clinical scores and a prolonged survival time accompanied with decreased viral RNA, protein expression and virus titer. The ingestion of honeysuckle attenuates EV71 replication and related pathogenesis partially through the upregulation of let-7a expression both in vitro and in vivo. Our previous report and the current findings imply that both honeysuckle and upregulated let-7a can execute a suppressive function against the replication of DENV and EV71. Taken together, this evidence indicates that honeysuckle can induce the expression of let-7a and that this miRNA as well as 11 other miRNAs have great potential to prevent and suppress EV71 replication.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Lonicera/chemistry , MicroRNAs/metabolism , Plant Extracts/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Enterovirus A, Human/physiology , Enterovirus Infections/drug therapy , Humans , Mice , Mice, Inbred ICR
20.
Antiviral Res ; 187: 105021, 2021 03.
Article in English | MEDLINE | ID: mdl-33508330

ABSTRACT

Cathelicidin antimicrobial peptides (human LL-37 and mouse CRAMP) are mainly virucidal to enveloped virus. However, the effects and relative mechanisms of LL-37 and CRAMP on non-enveloped virus are elusive. We herein found that CRAMP expression was significantly up-regulated post non-enveloped Enterovirus 71 (EV71) infection in different tissues of newborn ICR mice, while EV71 replication gradually declined post CRAMP up-regulation, indicating the antiviral potential of cathelicidin against EV71. In vitro antiviral assay showed that LL-37 and CRAMP markedly reduced cytopathic effects (CPE), intracellular viral RNA copy numbers, viral VP1 protein levels, and extracellular virons in U251 cells post EV71 infection, indicating that LL-37 and CRAMP significantly inhibited EV71 replication. Mechanism of action assay showed that LL-37 and CRAMP were not virucidal to EV71, but markedly regulated antiviral immune response in U251 cells. Co-incubation of LL-37 or CRAMP with U251 cells markedly increased the basal interferon-ß (IFN-ß) expression and interferon regulatory transcription factor 3 (IRF3) phosphorylation, modestly enhanced IFN-ß production and IRF3 phosphorylation upon EV71 infection, and significantly reduced interleukin-6 (IL-6) production and p38 mitogen-activated protein kinase (MAPK) activation post EV71 infection. Additionally, LL-37 and CRAMP directly inhibited viral binding to U251 cells. Collectively, LL-37 and CRAMP markedly inhibited EV71 replication via regulating antiviral response and inhibiting viral binding, providing potent candidates for peptide drug development against EV71 infection.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Enterovirus A, Human/drug effects , Immunity , Virus Attachment/drug effects , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Enterovirus A, Human/genetics , Enterovirus Infections/drug therapy , Enterovirus Infections/virology , Host Microbial Interactions , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred ICR , Phosphorylation , RNA, Viral , Up-Regulation , Vero Cells , Virus Replication/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL
...