Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
BMC Vet Res ; 20(1): 187, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730463

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV), a type of coronavirus, is one of the main pathogens that can infect pigs of all ages. It causes diarrhea and acute death of newborn piglets, resulting in massive economic losses to the worldwide swine industry. While vaccination remains the primary approach in combating PEDV, it often fails to address all the challenges posed by the infection, particularly in light of the emergence of evolving mutant strains. Therefore, there is a critical need to identify potent antiviral drugs that can effectively safeguard pigs against PEDV infection. RESULTS: In this study, the antiviral efficacy of SP2509, a specific antagonist of Lysine-specific demethylase 1(LSD1), was evaluated in vitro. The RT-qPCR, Western blot, TCID50, and IFA showed that at a concentration of 1µmol/L, SP2509 significantly inhibited PEDV infection. Additionally, viral life cycle assays showed that SP2509 operates by impeding PEDV internalization and replication rather than attachment and release. Regarding mechanism, in Huh-7 cells, knockdowns LSD1 can suppress PEDV replication. This indicated that the inhibition effect of SP2509 on PEDV largely depends on the activity of its target protein, LSD1. CONCLUSION: Our results in vitro show that SP2509 can inhibit PEDV infection during the internalization and replication stage and revealed a role of LSD1 as a restriction factor for PEDV. These imply that LSD1 might be a target for interfering with the viral infection, and SP2509 could be developed as an effective anti-PEDV agent.


Subject(s)
Antiviral Agents , Histone Demethylases , Porcine epidemic diarrhea virus , Virus Replication , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/pharmacology , Virus Replication/drug effects , Histone Demethylases/antagonists & inhibitors , Swine , Chlorocebus aethiops , Swine Diseases/virology , Swine Diseases/drug therapy , Coronavirus Infections/veterinary , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Vero Cells
2.
Virol J ; 21(1): 120, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816738

ABSTRACT

BACKGROUND: The Porcine Epidemic Diarrhea Virus (PEDV) has caused significant economic losses in the global swine industry. As a potential drug for treating diarrhea, the antiviral properties of attapulgite deserve further study. METHODS: In this study, various methods such as RT-qPCR, Western blot, viral titer assay, Cytopathic Effect, immunofluorescence analysis and transmission electron microscopy were used to detect the antiviral activity of attapulgite and to assess its inhibitory effect on PEDV. RESULTS: When exposed to the same amount of virus, there was a significant decrease in the expression of the S protein, resulting in a viral titer reduction from 10-5.613 TCID50/mL to 10-2.90 TCID50/mL, which represents a decrease of approximately 102.6 folds. Results of cytopathic effect and indirect immunofluorescence also indicate a notable decrease in viral infectivity after attapulgite treatment. Additionally, it was observed that modified materials after acidification had weaker antiviral efficacy compared to powdered samples that underwent ultrasonic disintegration, which showed the strongest antiviral effects. CONCLUSION: As a result, Attapulgite powders can trap and adsorb viruses to inhibit PEDV in vitro, leading to loss of viral infectivity. This study provides new materials for the development of novel disinfectants and antiviral additives.


Subject(s)
Antiviral Agents , Porcine epidemic diarrhea virus , Silicon Compounds , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/physiology , Animals , Antiviral Agents/pharmacology , Silicon Compounds/pharmacology , Silicon Compounds/chemistry , Chlorocebus aethiops , Magnesium Compounds/pharmacology , Swine , Vero Cells , Viral Load/drug effects , Cytopathogenic Effect, Viral/drug effects , Swine Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Microscopy, Electron, Transmission
3.
Int J Biol Macromol ; 270(Pt 2): 132408, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754683

ABSTRACT

Porcine Epidemic Diarrhea Virus (PEDV) is a highly contagious virus that causes Porcine Epidemic Diarrhea (PED). This enteric disease results in high mortality rates in piglets, leading to significant financial losses in the pig industry. However, vaccines cannot provide sufficient protection against epidemic strains. Spike (S) protein exposed on the surface of virion mediates PEDV entry into cells. Our findings imply that matrine (MT), a naturally occurring alkaloid, inhibits PEDV infection targeting S protein of virions and biological process of cells. The GLY434 residue in the autodocking site of the S protein and MT conserved based on sequence comparison. This study provides a comprehensive analysis of viral attachment, entry, and virucidal effects to investigate how that MT inhibits virus replication. MT inhibits PEDV attachment and entry by targeting S protein. MT was added to cells before, during, or after infection, it exhibits anti-PEDV activities and viricidal effects. Network pharmacology focuses on addressing causal mechanisms rather than just treating symptoms. We identified the key genes and screened the cell apoptosis involved in the inhibition of MT on PEDV infection in network pharmacology. MT significantly promotes cell apoptosis in PEDV-infected cells to inhibit PEDV infection by activating the MAPK signaling pathway. Collectively, we provide the biological foundations for the development of single components of traditional Chinese medicine to inhibit PEDV infection and spread.


Subject(s)
Alkaloids , Antiviral Agents , Apoptosis , MAP Kinase Signaling System , Matrines , Porcine epidemic diarrhea virus , Quinolizines , Spike Glycoprotein, Coronavirus , Quinolizines/pharmacology , Quinolizines/chemistry , Alkaloids/pharmacology , Alkaloids/chemistry , Animals , Porcine epidemic diarrhea virus/drug effects , Apoptosis/drug effects , Spike Glycoprotein, Coronavirus/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , MAP Kinase Signaling System/drug effects , Chlorocebus aethiops , Vero Cells , Swine , Virus Replication/drug effects , Virus Internalization/drug effects
4.
Virus Res ; 339: 199260, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37923169

ABSTRACT

Porcine epidemic diarrhea (PED) is a contagious intestinal disease caused by α-coronavirus porcine epidemic diarrhea virus (PEDV). At present, no effective vaccine is available to prevent the disease. Therefore, research for novel antivirals is important. This study aimed to identify the antiviral mechanism of Veratramine (VAM), which actively inhibits PEDV replication with a 50 % inhibitory concentration (IC50) of ∼5 µM. Upon VAM treatment, both PEDV-nucleocapsid (N) protein level and virus titer decreased significantly. The time-of-addition assay results showed that VAM could inhibit PEDV replication by blocking viral entry. Importantly, VAM could inhibit PEDV-induced phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) activity and further suppress micropinocytosis, which is required for PEDV entry. In addition, PI3K inhibitor LY294002 showed anti-PEDV activity by blocking viral entry as well. Taken together, VAM possessed anti-PEDV properties against the entry stage of PEDV by inhibiting the macropinocytosis pathway by suppressing the PI3K/Akt pathway. VAM could be considered as a lead compound for the development of anti-PEDV drugs and may be used during the viral entry stage of PEDV infection.


Subject(s)
Coronavirus Infections , Phosphatidylinositol 3-Kinases , Porcine epidemic diarrhea virus , Swine Diseases , Veratrum Alkaloids , Virus Internalization , Animals , Chlorocebus aethiops , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Phosphatidylinositol 3-Kinases/metabolism , Porcine epidemic diarrhea virus/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Swine , Swine Diseases/drug therapy , Veratrum Alkaloids/metabolism , Veratrum Alkaloids/pharmacology , Vero Cells , Virus Internalization/drug effects
5.
Viruses ; 14(2)2022 01 27.
Article in English | MEDLINE | ID: mdl-35215851

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) variant strains adversely affect the production of pigs globally. Vaccines derived from PEDV traditional strains impart less protection against the variant strains. Moreover, sequence diversity among different PEDV variant strains is also complicated. This necessitates developing alternative antiviral strategies for defending against PEDV. This study explored a natural product, Levistolide A (LA), to possess antiviral activity against PEDV. LA was found to suppress PEDV replication in a dose-dependent manner. And the inhibitory effect of LA against PEDV was maintained in the course of time. In terms of viral RNA and protein production, LA also showed a strong inhibitory effect. In addition, LA was indicated to inhibit PEDV from attaching to the cellular membrane or penetrating the cells. Further study revealed that LA can induce the generation of reactive oxygen species (ROS), and the corresponding inhibitor, NAC, was found to antagonize the effect of LA on inhibiting PEDV replication. This illustrated that the LA-induced ROS generation played an important role in its anti-PEDV activity. LA was also identified to stimulate ER stress, which is an important consequence of ROS production and was proven to be able to inhibit PEDV replication. To conclude, this study revealed that LA can inhibit PEDV replication via inducing ROS generation.


Subject(s)
Antiviral Agents/pharmacology , Benzofurans/pharmacology , Porcine epidemic diarrhea virus/drug effects , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Endoplasmic Reticulum Stress , Porcine epidemic diarrhea virus/physiology , RNA, Viral/metabolism , Sus scrofa , Vero Cells , Viral Proteins/metabolism , Virus Replication/drug effects
6.
Viruses ; 14(2)2022 02 15.
Article in English | MEDLINE | ID: mdl-35215995

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is an alphacoronavirus that causes severe watery diarrhea in piglets with high morbidity and mortality, resulting in serious economic losses to the farming industry. Ergosterol peroxide (EP) is a sterol with diverse biological activities including antiviral activity. In this study, we explored whether EP extracted from the fruiting body of the mushroom Cryptoporus volvatus had the potential to inhibit PEDV infection in Vero cells. The results revealed that EP had a remarkable inhibitory effect on PEDV infection. It could significantly inhibit multiple stages of the PEDV life cycle, including internalization, replication and release, and could directly inactivate PDCoV infectivity. However, it did not affect PEDV attachment. Furthermore, EP alleviated PEDV-induced apoptosis and mitigated the decrease in mitochondrial membrane potential caused by PEDV infection. It suppressed ROS generation and p53 activation caused by PEDV infection. The ROS scavenger N-acetyl-l-cysteine (NAC) and the p53 specific inhibitor Pifithrin-α (PFT-α) suppressed PEDV-induced apoptosis and impeded viral replication, suggesting that ROS and p53 play an important role in PEDV-induced apoptosis and viral replication. Collectively, EP can prevent PEDV internalization, replication and release, possesses the ability to directly inactivate PEDV, and can inhibit PEDV-induced apoptosis by interfering with PEDV-induced ROS production and p53 activation. These findings highlight the therapeutic potential of EP against PEDV infection.


Subject(s)
Coronavirus Infections/veterinary , Ergosterol/analogs & derivatives , Porcine epidemic diarrhea virus/drug effects , Reactive Oxygen Species/metabolism , Swine Diseases/virology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Chlorocebus aethiops , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Ergosterol/pharmacology , Polyporaceae/chemistry , Swine , Swine Diseases/drug therapy , Vero Cells , Virus Replication/drug effects
7.
Viruses ; 13(12)2021 12 16.
Article in English | MEDLINE | ID: mdl-34960796

ABSTRACT

Porcine epidemic diarrhea virus (PEDV), an enteric coronavirus, causes neonatal pig acute gastrointestinal infection with a characterization of severe diarrhea, vomiting, high morbidity, and high mortality, resulting in tremendous damages to the swine industry. Neither specific antiviral drugs nor effective vaccines are available, posing a high priority to screen antiviral drugs. The aim of this study is to investigate anti-PEDV effects of carbazole alkaloid derivatives. Eighteen carbazole derivatives (No.1 to No.18) were synthesized, and No.5, No.7, and No.18 were identified to markedly reduce the replication of enhanced green fluorescent protein (EGFP) inserted-PEDV, and the mRNA level of PEDV N. Flow cytometry assay, coupled with CCK8 assay, confirmed No.7 and No.18 carbazole derivatives displayed high inhibition effects with low cell toxicity. Furthermore, time course analysis indicated No.7 and No.18 carbazole derivatives exerted inhibition at the early stage of the viral life cycle. Collectively, the analysis underlines the benefit of carbazole derivatives as potential inhibitors of PEDV, and provides candidates for the development of novel therapeutic agents.


Subject(s)
Antiviral Agents/pharmacology , Carbazoles/pharmacology , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/chemistry , Carbazoles/chemistry , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Molecular Structure , Vero Cells , Virus Attachment/drug effects , Virus Replication/drug effects
8.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578406

ABSTRACT

The porcine epidemic diarrhea virus (PEDV) is an Alphacoronavirus (α-CoV) that causes high mortality in infected piglets, resulting in serious economic losses in the farming industry. Hypericin is a dianthrone compound that has been shown as an antiviral activity on several viruses. Here, we first evaluated the antiviral effect of hypericin in PEDV and found the viral replication and egression were significantly reduced with hypericin post-treatment. As hypericin has been shown in SARS-CoV-2 that it is bound to viral 3CLpro, we thus established a molecular docking between hypericin and PEDV 3CLpro using different software and found hypericin bound to 3CLpro through two pockets. These binding pockets were further verified by another docking between hypericin and PEDV 3CLpro pocket mutants, and the fluorescence resonance energy transfer (FRET) assay confirmed that hypericin inhibits the PEDV 3CLpro activity. Moreover, the alignments of α-CoV 3CLpro sequences or crystal structure revealed that the pockets mediating hypericin and PEDV 3CLpro binding were highly conserved, especially in transmissible gastroenteritis virus (TGEV). We then validated the anti-TGEV effect of hypericin through viral replication and egression. Overall, our results push forward that hypericin was for the first time shown to have an inhibitory effect on PEDV and TGEV by targeting 3CLpro, and it deserves further attention as not only a pan-anti-α-CoV compound but potentially also as a compound of other coronaviral infections.


Subject(s)
Alphacoronavirus/drug effects , Alphacoronavirus/physiology , Anthracenes/pharmacology , Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus Infections/virology , Perylene/analogs & derivatives , Virus Replication/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Coronavirus 3C Proteases/chemistry , Enzyme Activation/drug effects , Models, Molecular , Perylene/pharmacology , Porcine epidemic diarrhea virus/drug effects , Recombinant Proteins , Structure-Activity Relationship , Swine , Swine Diseases/virology , Vero Cells
9.
Viruses ; 13(9)2021 09 18.
Article in English | MEDLINE | ID: mdl-34578449

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is the predominant cause of an acute, highly contagious enteric disease in neonatal piglets. There are currently no approved drugs against PEDV infection. Here, we report the development of a nanoluciferase (NLuc)-based high-throughput screening (HTS) platform to identify novel anti-PEDV compounds. We constructed a full-length cDNA clone for a cell-adapted PEDV strain YN150. Using reverse genetics, we replaced the open reading frame 3 (ORF3) in the viral genome with an NLuc gene to engineer a recombinant PEDV expressing NLuc (rPEDV-NLuc). rPEDV-NLuc produced similar plaque morphology and showed similar growth kinetics compared with the wild-type PEDV in vitro. Remarkably, the level of luciferase activity could be stably detected in rPEDV-NLuc-infected cells and exhibited a strong positive correlation with the viral titers. Given that NLuc expression represents a direct readout of PEDV replication, anti-PEDV compounds could be easily identified by quantifying the NLuc activity. Using this platform, we screened for the anti-PEDV compounds from a library of 803 natural products and identified 25 compounds that could significantly inhibit PEDV replication. Interestingly, 7 of the 25 identified compounds were natural antioxidants, including Betulonic acid, Ursonic acid, esculetin, lithocholic acid, nordihydroguaiaretic acid, caffeic acid phenethyl ester, and grape seed extract. As expected, all of the antioxidants could potently reduce PEDV-induced oxygen species production, which, in turn, inhibit PEDV replication in a dose-dependent manner. Collectively, our findings provide a powerful platform for the rapid screening of promising therapeutic compounds against PEDV infection.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , High-Throughput Screening Assays , Luciferases/genetics , Microbial Sensitivity Tests , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/genetics , Animals , Antioxidants/pharmacology , Chlorocebus aethiops , Genetic Engineering , Genome, Viral , Luciferases/metabolism , Reactive Oxygen Species/metabolism , Reverse Genetics , Vero Cells
10.
Oxid Med Cell Longev ; 2021: 3055810, 2021.
Article in English | MEDLINE | ID: mdl-34257799

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) has reemerged throughout the world in the past ten years and caused huge economic losses to the swine industry. No drugs are available to prevent or treat PEDV infection in piglets. Zinc oxide (ZnO) has been shown to reduce diarrhea. However, little is known about its role in PEDV infection. In this study, twenty-four 7-day-old piglets were randomly divided into three treatment groups: control, PEDV, and ZnO+PEDV. Piglets in the ZnO+PEDV group were orally administered with 100 mg/kg·BW ZnO and then inoculated PEDV at a dose of 104.5 TCID50 (50% tissue culture infectious dose) per pig. Growth performance, histologic lesions, viral load, indicators of intestinal damage, inflammation, and oxidative stress were recorded or detected to determine the effect of ZnO on PEDV infection. And the underlying mechanisms were revealed by microarray and proteomic analyses. Results showed that ZnO administration mitigated diarrhea and the reduction of average daily weight gain induced by PEDV infection. ZnO could inhibit PEDV replication in the small intestine and colon. Both villus height and crypt depth were affected by PEDV infection in the duodenum and jejunum, which could be rescued by ZnO administration. Moreover, the activity of catalase was decreased both in plasma and intestine after PEDV infection, while increased in the intestine by ZnO administration. PEDV infection also significantly increased the concentration of H2O2 in jejunal and ileum and decreased the activity of total superoxide dismutase and glutathione peroxidase in plasma, whereas ZnO administration obviously increased the activity of total superoxide dismutase and decreased the concentration of H2O2 in the ileum. The concentrations of IL-1ß, IL-6, and IL-8 in the plasma were all decreased upon ZnO administration. A large number of differentially expressed genes and proteins were identified in the ileum among the three groups by microarray and proteomic analyses. Gene Ontology and Reactome pathway analyses indicated that neutrophil degranulation and nutrient metabolism were the main biological process and pathways in both PEDV infection and ZnO administration. Overall, ZnO administration could improve growth performance, intestinal redox status, morphology, and function and reduce diarrhea in PEDV-infected piglets; ZnO could exert antiviral and anti-inflammatory effects on PEDV-infected piglets probably through regulating neutrophil degranulation. Our findings have important implications in piglet and infant nutrition.


Subject(s)
Dermatologic Agents/therapeutic use , Neutrophils/drug effects , Porcine epidemic diarrhea virus/drug effects , Zinc Oxide/therapeutic use , Animals , Dermatologic Agents/pharmacology , Swine , Zinc Oxide/pharmacology
11.
Res Vet Sci ; 139: 32-42, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34246941

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is an enteropathogenic coronavirus; it causes diarrhea in pigs and is associated with high morbidity and mortality in sucking piglets. In this study, we performed in vitro and in vivo experiments to determine the inhibitory effects of Lactobacillus plantarum metabolites (LPM) on PEDV replication. Gas chromatography-mass spectrometry revealed exopolysaccharides to be one of the main components of LPM. We then determine whether L. plantarum exopolysaccharides (LPE) have an antiviral effect and also detected the expression levels of the apoptosis-related genes Bax and Bcl-2 and of the pro-apoptotic protein caspase-3. Further, we assessed the transcription levels of an immune-related protein (STAT1) and antiviral factors (MX1, MX2, ISG15, ZAP, PKR, and OAS1). Our results showed that the most effective method was to pretreat cells with LPM and that the optimal dose of LPM that could be safely administered to Vero cells was 1/8 times of the stock solution. LPE had a strong inhibitory effect on PEDV; the most effective method of administration was to co-incubate cells with LPE and PEDV, and the optimal concentration of LPE was 1.35 mg/mL. To conclude, LPE prevented PEDV adsorption and also alleviated inflammatory responses and induced early apoptosis of injured cells, but it could not regulate the immune function of cells.


Subject(s)
Lactobacillus plantarum/metabolism , Polysaccharides/pharmacology , Polysaccharides/therapeutic use , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/growth & development , Swine Diseases/drug therapy , Swine Diseases/virology , Virus Replication/drug effects , Animals , Apoptosis/drug effects , Chlorocebus aethiops , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Diarrhea/drug therapy , Diarrhea/veterinary , Diarrhea/virology , Inflammation/drug therapy , Porcine epidemic diarrhea virus/immunology , Swine , Swine Diseases/immunology , Vero Cells , Virus Attachment/drug effects
12.
J Virol ; 95(19): e0064521, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34287043

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a globally distributed alphacoronavirus that has reemerged lately, resulting in large economic losses. During viral infection, type I interferon (IFN-I) plays a vital role in the antiviral innate immunity. However, PEDV has evolved strategies to limit IFN-I production. To suppress virus replication, the host must activate IFN-stimulated genes and some host restriction factors to circumvent viral replication. This study observed that PEDV infection induced early growth response gene 1 (EGR1) expression in PEDV-permissive cells. EGR1 overexpression remarkably suppressed PEDV replication. In contrast, depletion of EGR1 led to a significant increase in viral replication. EGR1 suppressed PEDV replication by directly binding to the IFN-regulated antiviral (IRAV) promoter and upregulating IRAV expression. A detailed analysis revealed that IRAV interacts and colocalizes with the PEDV nucleocapsid (N) protein, inducing N protein degradation via the E3 ubiquitin ligase MARCH8 to catalyze N protein ubiquitination. Knockdown of endogenous MARCH8 significantly reversed IRAV-mediated N protein degradation. The collective findings demonstrate a new mechanism of EGR1-mediated viral restriction, in which EGR1 upregulates the expression of IRAV to degrade PEDV N protein through MARCH8. IMPORTANCE PEDV is a highly contagious enteric coronavirus that has rapidly emerged worldwide and has caused severe economic losses. No currently available drugs or vaccines can effectively control PEDV. PEDV has evolved many strategies to limit IFN-I production. We identified EGR1 as a novel host restriction factor and demonstrated that EGR1 suppresses PEDV replication by directly binding to the IRAV promoter and upregulating the expression of IRAV, which interacts with and degrades the PEDV N protein via the E3 ubiquitin ligase MARCH8 to catalyze nucleocapsid protein ubiquitination, which adds another layer of complexity to the innate antiviral immunity of this newly identified restriction factor. A better understanding of the innate immune response to PEDV infection will aid the development of novel therapeutic targets and more effective vaccines against virus infection.


Subject(s)
Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/pharmacology , Nucleocapsid Proteins/metabolism , Porcine epidemic diarrhea virus/drug effects , RNA-Binding Proteins/metabolism , Virus Replication/drug effects , Animals , Antiviral Agents/metabolism , Chlorocebus aethiops , Coronavirus Infections , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Type I/metabolism , Nucleocapsid/metabolism , Porcine epidemic diarrhea virus/genetics , Swine , Swine Diseases/virology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vero Cells
13.
J Virol ; 95(16): e0018721, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34037422

ABSTRACT

Subversion of the host cell cycle to facilitate viral replication is a common feature of coronavirus infections. Coronavirus nucleocapsid (N) protein can modulate the host cell cycle, but the mechanistic details remain largely unknown. Here, we investigated the effects of manipulation of porcine epidemic diarrhea virus (PEDV) N protein on the cell cycle and the influence on viral replication. Results indicated that PEDV N induced Vero E6 cell cycle arrest at S-phase, which promoted viral replication (P < 0.05). S-phase arrest was dependent on the N protein nuclear localization signal S71NWHFYYLGTGPHADLRYRT90 and the interaction between N protein and p53. In the nucleus, the binding of N protein to p53 maintained consistently high-level expression of p53, which activated the p53-DREAM pathway. The key domain of the N protein interacting with p53 was revealed to be S171RGNSQNRGNNQGRGASQNRGGNN194 (NS171-N194), in which G183RG185 are core residues. NS171-N194 and G183RG185 were essential for N-induced S-phase arrest. Moreover, small molecular drugs targeting the NS171-N194 domain of the PEDV N protein were screened through molecular docking. Hyperoside could antagonize N protein-induced S-phase arrest by interfering with interaction between N protein and p53 and inhibit viral replication (P < 0.05). The above-described experiments were also validated in porcine intestinal cells, and data were in line with results in Vero E6 cells. Therefore, these results reveal the PEDV N protein interacts with p53 to activate the p53-DREAM pathway, and subsequently induces S-phase arrest to create a favorable environment for virus replication. These findings provide new insight into the PEDV-host interaction and the design of novel antiviral strategies against PEDV. IMPORTANCE Many viruses subvert the host cell cycle to create a cellular environment that promotes viral growth. PEDV, an emerging and reemerging coronavirus, has led to substantial economic loss in the global swine industry. Our study is the first to demonstrate that PEDV N-induced cell cycle arrest during the S-phase promotes viral replication. We identified a novel mechanism of PEDV N-induced S-phase arrest, where the binding of PEDV N protein to p53 maintains consistently high levels of p53 expression in the nucleus to mediate S-phase arrest by activating the p53-DREAM pathway. Furthermore, a small molecular compound, hyperoside, targeted the PEDV N protein, interfering with the interaction between the N protein and p53 and, importantly, inhibited PEDV replication by antagonizing cell cycle arrest. This study reveals a new mechanism of PEDV-host interaction and also provides a novel antiviral strategy for PEDV. These data provide a foundation for further research into coronavirus-host interactions.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Nucleocapsid Proteins/chemistry , Host-Pathogen Interactions/drug effects , Porcine epidemic diarrhea virus/drug effects , Quercetin/analogs & derivatives , Tumor Suppressor Protein p53/chemistry , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Binding Sites , Cell Line , Chlorocebus aethiops , Coronavirus Infections/drug therapy , Coronavirus Infections/genetics , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins/antagonists & inhibitors , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , Gene Expression Regulation , High-Throughput Screening Assays , Host-Pathogen Interactions/genetics , Molecular Docking Simulation , Nuclear Localization Signals , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Quercetin/chemistry , Quercetin/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , S Phase Cell Cycle Checkpoints/genetics , Signal Transduction , Swine , Swine Diseases/drug therapy , Swine Diseases/genetics , Swine Diseases/metabolism , Swine Diseases/virology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vero Cells , Virus Replication/drug effects
14.
Viruses ; 13(4)2021 03 30.
Article in English | MEDLINE | ID: mdl-33808275

ABSTRACT

Porcine epidemic diarrhea virus (PEDV), an enteropathogenic coronavirus, has catastrophic impacts on the global pig industry. Owing to the lack of effective vaccines and specific therapeutic options for PEDV, it is pertinent to develop new and available antivirals. This study identified, for the first time, a salinomycin that actively inhibited PEDV replication in Vero cells in a dose-dependent manner. Furthermore, salinomycin significantly inhibited PEDV infection by suppressing the entry and post-entry of PEDV in Vero cells. It did not directly interact with or inactivate PEDV particles, but it significantly ameliorated the activation of Erk1/2, JNK and p38MAPK signaling pathways that are associated with PEDV infection. This implied that salinomycin inhibits PEDV replication by altering MAPK pathway activation. Notably, the PEDV induced increase in reactive oxidative species (ROS) was not decreased, indicating that salinomycin suppresses PEDV replication through a pathway that is an independent pathway of viral-induced ROS. Therefore, salinomycin is a potential drug that can be used for treating PEDV infection.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/drug effects , Pyrans/pharmacology , Swine Diseases/virology , Animals , Chlorocebus aethiops , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Coronavirus Infections/virology , MAP Kinase Signaling System , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Swine , Swine Diseases/drug therapy , Swine Diseases/genetics , Swine Diseases/metabolism , Vero Cells , Virus Replication/drug effects
15.
Sci Rep ; 11(1): 6552, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753826

ABSTRACT

Puerarin has been reported to be an excellent antioxidant, anti-inflammatory and antimicrobial agent, but the potential effect of puerarin on porcine epidemic diarrhea virus (PEDV) is unclear. This study aimed to determine whether puerarin could alleviate intestinal injury in piglets infected with PEDV. A PEDV (Yunnan province strain) infection model was applied to 7-day-old piglets at 104.5 TCID50 (50% tissue culture infectious dose). Piglets were orally administered with puerarin at the dosage of 0.5 mg/kg body weight from day 5 to day 9. On day 9 of the trial, piglets were inoculated orally with PEDV. Three days later, jugular vein blood and intestinal samples were collected. Results showed puerarin reduced morbidity of piglets infected with PEDV. In addition, puerarin reduced the activities of aspartate aminotransferase and alkaline phosphatase, the ratio of serum aspartate aminotransferase to serum alanine aminotransferase, the number of white blood cells and neutrophils, and the plasma concentrations of interleukin-6, interleukin-8 and tumor necrosis factor-α, as well as protein abundances of heat shock protein-70 in PEDV-infected piglets. Moreover, puerarin increased D-xylose concentration but decreased intestinal fatty acid-binding protein concentration and diamine oxidase activity in the plasma of piglets infected with PEDV. Puerarin increased the activities of total superoxide dismutase, glutathione peroxidase and catalase, while decreasing the activities of myeloperoxidase and concentration of hydrogen peroxide in both the intestine and plasma of PEDV-infected piglets. Puerarin decreased mRNA levels of glutathione S-transferase omega 2 but increased the levels of nuclear factor erythroid 2-related factor 2. Furthermore, puerarin increased the abundance of total eubacteria (16S rRNA), Enterococcus genus, Lactobacillus genus and Enterobacteriaceae family in the intestine, but reduced the abundance of Clostridium coccoides in the caecum. These data indicate puerarin improved intestinal function in piglets infected by PEDV and may be a promising supplement for the prevention of PEDV infection.


Subject(s)
Intestines/drug effects , Intestines/virology , Isoflavones/pharmacology , Porcine epidemic diarrhea virus/drug effects , Swine Diseases/virology , Animals , Animals, Newborn , Antioxidants/metabolism , Biomarkers , Cytokines/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Intestines/pathology , Oxidation-Reduction , Oxidative Stress , Swine , Swine Diseases/metabolism , Swine Diseases/pathology
16.
J Nat Prod ; 84(4): 1175-1184, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33760626

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) has become increasingly problematic around the world, not only for its hazards to livestock but also due to the possibility that it is a zoonotic disease. Although vaccine therapy has made some progress toward PEDV control, additional effective therapeutic strategies against PEDV are needed, such as the development of chemotherapeutic agents. The aim of this work was to identify novel anti-PEDV agents by designing and synthesizing a series of phenanthridine derivatives. Among them, three compounds (compounds 1, 2, and 4) were identified as potent anti-PEDV agents exhibiting suppression of host cell heat shock cognate 70 (Hsc70) expression. Mechanism studies revealed that host Hsc70 is involved in the replication of PEDV, and its expression can be suppressed by destabilization of the mRNA, resulting in inhibition of PEDV replication. Activity against PEDV in vivo in PEDV-infected piglets suggested that phenanthridine derivatives are the first host-acting potential anti-PEDV agents.


Subject(s)
Antiviral Agents/pharmacology , HSC70 Heat-Shock Proteins/metabolism , Phenanthridines/pharmacology , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/chemical synthesis , Cell Line , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Drug Design , Molecular Structure , Phenanthridines/chemical synthesis , Swine
17.
Virol Sin ; 36(5): 997-1005, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33751399

ABSTRACT

Porcine epidemic diarrhea (PED) caused by porcine epidemic diarrhea virus (PEDV) is widespread in the world. In recent years, the increased virulence of the virus due to viral variations, has caused great economic losses to the pig industry in many countries. It is always worthy to find effective therapeutic methods for PED. As an important class of antivirals, nucleoside drugs which target viral polymerases have been applied in treating human viral infections for half a century. Herein, we evaluated the anti-PEDV potential of three broad-spectrum antiviral nucleoside analogs, remdesivir (RDV), its parent nucleoside (RDV-N) and ß-D-N4-hydroxycytidine (NHC). Among them, RDV-N was the most active agent in Vero E6 cells with EC50 of 0.31 µmol/L, and more potent than RDV (EC50 = 0.74 µmol/L) and NHC (EC50 = 1.17 µmol/L). The activity of RDV-N was further confirmed using an indirect immuno-fluorescence assay. Moreover, RDV-N exhibited a good safety profile in cells and in mice. The high sequence similarity of the polymerase functional domains of PEDV with other five porcine coronaviruses indicated a broader antiviral spectrum for the three compounds. Generally, RDV-N is a promising broad-spectrum antiviral nucleoside, and it would be worthy to make some structural modifications to increase its oral bioavailability.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Coronavirus Infections , Cytidine/pharmacology , Porcine epidemic diarrhea virus , Swine Diseases , Adenosine Monophosphate/pharmacology , Alanine/pharmacology , Animals , Chlorocebus aethiops , Cytidine/analogs & derivatives , Mice , Nucleosides/pharmacology , Parents , Porcine epidemic diarrhea virus/drug effects , Swine , Vero Cells
18.
Vet Microbiol ; 254: 108994, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33486326

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) encodes many multifunctional proteins that inhibit host innate immune response during virus infection. As one of important structural proteins, PEDV E protein has been found to block the production of type I interferon (IFN) in virus life cycle, but little is known about this process that E protein subverts host innate immune. Thus, in this present study, we initiated the construction of eukaryotic expression vectors to express PEDV E protein. Subsequently, cellular localization analysis was performed and the results showed that the majority of PEDV E protein distributed at cytoplasm and localized in endoplasmic reticulum (ER). Over-expression of PEDV E protein significantly inhibited poly(I:C)-induced IFN-ß and IFN-stimulated genes (ISGs) productions. We also found that PEDV E protein remarkably suppressed the protein expression of RIG-I signaling-associated molecules, but all their corresponding mRNA levels remained unaffected and unchanged. Furthermore, PEDV E protein obviously interfered with the translocation of IRF3 from cytoplasm to nucleus through direct interaction with IRF3, which is crucial for the IFN-ß production induced by poly(I:C). Taken together, our results suggested that PEDV E protein acts as an IFN-ß antagonist through suppression of the RIG-I-mediated signaling. This study will pave the way for the further investigation into the molecular mechanisms by which PEDV E protein evades host innate immune response.


Subject(s)
DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/immunology , Interferon-beta/immunology , Porcine epidemic diarrhea virus/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Viral Proteins/genetics , Animals , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Immune Evasion , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/antagonists & inhibitors , Interferon-beta/biosynthesis , Interferon-beta/genetics , Poly I-C/pharmacology , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Swine , Viral Proteins/metabolism
19.
Front Immunol ; 12: 797476, 2021.
Article in English | MEDLINE | ID: mdl-35095875

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) has reemerged as the main pathogen of piglets due to its high mutation feature. Monolaurin (ML) is a natural compound with a wide range of antibacterial and antiviral activities. However, the role of ML in PEDV infection is still unknown. This study aimed to evaluate the effect of ML on the growth performance, intestinal function, virus replication and cytokine response in piglets infected with PEDV, and to reveal the mechanism through proteomics analysis. Piglets were orally administrated with ML at a dose of 100 mg/kg·BW for 7 days before PEDV infection. Results showed that although there was no significant effect on the growth performance of piglets, ML administration alleviated the diarrhea caused by PEDV infection. ML administration promoted the recovery of intestinal villi, thereby improving intestinal function. Meanwhile, PEDV replication was significantly inhibited, and PEDV-induced expression of IL-6 and IL-8 were decreased with ML administration. Proteomics analyses showed that 38 proteins were differentially expressed between PEDV and ML+PEDV groups and were significantly enriched in the interferon-related pathways. This suggests ML could promote the restoration of homeostasis by regulating the interferon pathway. Overall, the present study demonstrated ML could confer a protective effect against PEDV infection in piglets and may be developed as a drug or feed additive to prevent and control PEDV disease.


Subject(s)
Coronavirus Infections/prevention & control , Interferons/metabolism , Laurates/pharmacology , Monoglycerides/pharmacology , Porcine epidemic diarrhea virus/drug effects , Signal Transduction/drug effects , Swine Diseases/prevention & control , Animals , Animals, Newborn , Chromatography, Liquid/methods , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Cytokines/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Laurates/administration & dosage , Monoglycerides/administration & dosage , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/physiology , Protective Agents/pharmacology , Proteome/metabolism , Proteomics/methods , Swine , Swine Diseases/metabolism , Swine Diseases/virology , Tandem Mass Spectrometry/methods , Viral Load/drug effects , Viral Load/genetics
20.
Int J Mol Sci ; 21(21)2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33142980

ABSTRACT

For the last decade, porcine epidemic diarrhea virus (PEDV) variant strains have caused severe damage to the global pig industry. Until now, no effective antivirals have been developed for the therapeutic treatment of PEDV infection. In the present study, we found that quercetin significantly suppressed PEDV infection at noncytotoxic concentrations. A molecular docking study indicated that quercetin might bind the active site and binding pocket of PEDV 3C-like protease (3CLpro). Surface plasmon resonance (SPR) analysis revealed that quercetin exhibited a binding affinity to PEDV 3CLpro. Based on the results of the fluorescence resonance energy transfer (FRET) assay, quercetin was proven to exert an inhibitory effect on PEDV 3CLpro. Since coronavirus 3CLpro is an important drug target and participates in the viral replication process, quercetin should be developed as a novel drug in the control of PEDV infection.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/drug effects , Quercetin/pharmacology , Swine Diseases/drug therapy , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication , Animals , Antiviral Agents/therapeutic use , Coronavirus 3C Proteases , Coronavirus Infections/drug therapy , Cysteine Endopeptidases , Porcine epidemic diarrhea virus/physiology , Quercetin/therapeutic use , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...