INTRODUCTION
Porcine reproductive and respiratory syndrome virus (PRRSV) is an enveloped, positive-sense single-stranded RNA virus with a genome size of 14.9–15.5 kb, and it belongs to the
Arteriviridae family (
1,
2). This virus causes reproductive disorders in sows and severe respiratory disease in swine, generating massive economic losses in the global pig farming industry (
3,
4). Since its emergence in the 1980s, PRRSV has undergone significant mutation and recombination (
5,
6). However, effective vaccines or drugs that prevent or control PRRSV have yet to be developed, and there is an urgent need for new effective anti-PRRSV drugs.
Hyperoside is a natural flavonoid that can be extracted from various plants, including
Cuscuta chinensis Lam.,
Forsythia suspensa, and
Crataegus pinnatifida Bge (
7). Previous studies have shown that hyperoside possesses a broad spectrum of biological activities, including anti-inflammatory, antiviral, antioxidant, antihyperglycemic, and anticancer activities (
8–11). In recent years, hyperoside has also been found to inhibit several human and animal viruses, such as herpes simplex virus type 1 (
12), hepatitis B virus (
13), infectious bronchitis virus (
14), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (
15), African swine fever virus (
16), porcine epidemic diarrhea virus (PEDV) (
10), and equine herpesvirus type 8 (EHV-8) (
17). However, information regarding the anti-PRRSV activity of hyperoside and the potential molecular mechanisms underlying its effects on this virus is limited.
Both inflammatory cytokines and autophagy play crucial roles in viral infection and pathogenesis (
18). PRRSV infection significantly increases the levels of inflammatory cytokines such as IL-1β, IL-6, IL-8, and TNF-α in porcine alveolar macrophages (PAMs) and swine (
19,
20), with uncontrolled inflammation aggravating lung damage (
21). Recent studies have shown that compounds that downregulate pro-inflammatory factors (
22), such as the itaconate derivative 4-OI22 and allicin (
23), can effectively inhibit PRRSV infection. Similarly, autophagy—an evolutionarily conserved cellular process that maintains homeostasis (
24–26)—can either promote or inhibit viral replication, depending on the virus type (
27,
28). Previous research has demonstrated that PRRSV-induced autophagy facilitates viral self-replication (
29,
30), while disrupting this autophagy response can suppress PRRSV infection (
31). Notably, hyperoside has been shown to inhibit inflammatory responses (
32) and suppress autophagy (
33,
34) in various disease models, although its effects on PRRSV-induced inflammation and autophagy remain unexplored.
In the present study, the anti-PRRSV activity of hyperoside was investigated, both in susceptible cells and in piglets. Moreover, the potential anti-PRRSV mechanisms of hyperoside were explored. Our results showed that hyperoside could effectively confer resistance against PRRSV in vitro at multiple replication stages. Further analysis revealed that hyperoside could also attenuate the overexpression of pro-inflammatory factors induced by PRRSV by suppressing the Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway. Additionally, our findings showed that hyperoside alleviated PRRSV-induced autophagy by activating the p62/Nrf2/Keap1 signaling pathway. Finally, we demonstrated that hyperoside was effective in reducing the PRRSV load and lung injury in piglets in vivo. Our data indicate that hyperoside may serve as a promising therapeutic agent against PRRSV.
MATERIALS AND METHODS
Cells, viruses, and reagents
MARC-145 cells were obtained from the China Center for Type Culture Collection (Wuhan, China) and maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and penicillin-streptomycin at 37°C under 5% CO2. PAMs were obtained from 8-week-old PRRSV-negative pigs as previously described and cultured in RPMI-1640 medium supplemented with 10% FBS.
The PRRSV-1 strains GZ11-G1 (GenBank: KF001144.1) and P073-3 (GenBank: MK214314.1) and the PRRSV-2 strains SD16 (GenBank: JX087437.1), CH-1a (GenBank: AY032626), JXA1 (GenBank: EF112445.1), and NADC30-like (GenBank: KX766379) were used in this study. These PRRSV isolates were allowed to proliferate in MARC-145 cells and titrated via a plaque formation assay, as described previously (
35,
36).
Hyperoside was obtained from Sigma?Aldrich (Saint Louis, USA) and dissolved in dimethylsulfoxide (DMSO). 3-Methyladenine (3-MA, an autophagy inhibitor) was obtained from Shandong Sparkjade Biotechnology Co., Ltd. (Jinan, China). Finally, porcine PRRSV convalescent serum, which contained monoclonal antibodies against the PRRSV-1 and PRRSV-2 N proteins, was prepared in our laboratory.
Cytotoxicity assay
The cytotoxicity of hyperoside to MARC-145 and PAM cells was examined as described in previous studies (
17). Briefly, MARC-145 and PAMs were preseeded into 96-well plates overnight and then treated with various concentrations of hyperoside (0, 15, 30, 60, 120, and 200 μM) for 24 h. Then, the medium was removed, and the cells were resuspended in 50 μL of DMEM or RPMI-1640 containing 10% Cell Counting Kit-8 (CCK-8; Shandong Sparkjade Biotechnology Co., Ltd., Jinan, China) and incubated at 37°C for 2 h. To calculate cell viability, the optical density (OD) of each well was measured at 450 nm via an Epoch microplate spectrophotometer (Biotek, USA) and analyzed via GraphPad Prism 8.0. DMSO was used as the negative control for these experiments.
RNA extraction and quantitative real-time polymerase chain reaction
All the cell samples were harvested at the indicated time points, and total RNA was extracted via SparkZol Reagent (Jinan, China); subsequently, 1 μg of RNA extracted from each group of cells was reverse transcribed into cDNA via the PrimeScript RT Master Mix (Takara, Japan). Then, quantitative PCR (qPCR) was performed to quantify target gene expression via the 2
???Ct method, as described previously (
37).
GAPDH transcripts were amplified to normalize the total RNA input. The primer information is listed in
Table 1.
Absolute quantification was also performed to evaluate the RNA genome copy numbers of PRRSV-1 and PRRSV-2 in the cell culture supernatant, as described previously (
35). Two recombinant plasmids, pMD18-T-N-1 and pMD18-T-N-2, containing the PRRSV-1
ORF7 gene and PRRSV-2
ORF7 gene, respectively, were used. These plasmids served as templates for qPCR at the indicated times.
Western blot analysis
The cells were collected at the indicated time points and lysed with 2× Laemmli sample buffer. The lysates were then boiled, and equal amounts of samples were separated via 10% SDS-PAGE and transferred onto polyvinylidene fluoride (PVDF) membranes as described previously (
38,
39). The PVDF membranes were further blocked with 5% serum albumin and incubated with specific antibodies to detect different target proteins. α-Tubulin served as the loading control. Finally, the membranes were imaged via a ChemiDoc XRS imaging system (Bio-Rad, USA). The primary antibodies used in the present study included antibodies against TLR4, NF-kB p65, p-NF-kB p65, IkB, p-IkB, and LC3 (Cell Signaling Technology, USA); anti-p62 and anti-Nrf2 antibodies (Servicebio, Wuhan, China); and anti-α-Tubulin and anti-Keap1 antibodies (Abcam, Cambridge, UK). Horseradish peroxidase-conjugated anti-mouse IgG and anti-rabbit IgG were purchased from Thermo Fisher Scientific (Massachusetts, USA).
Viral titration
MARC-145 cells were grown to approximately 80%–90% confluency in 96-well plates. The viral suspensions were serially diluted 10-fold, and 100 μL of each dilution was added to the wells (
n = 8) and incubated for 1 h. Subsequently, the cells were incubated with DMEM containing 3% FBS for 5 days. The 50% tissue culture infectious dose (TCID
50) was calculated via the Reed-Muench method according to previous reports (
36) and analyzed via GraphPad Prism 8.0.
Antiviral activity assay
MARC-145 cells and PAMs were seeded into 12-well plates and cultured until they reached 90% confluence. These cells were subsequently treated with various concentrations of hyperoside (15, 30, 60, or 120 μM) or the indicated medium containing 0.1% DMSO for 2 h. Following incubation, the cells were infected with PRRSV SD16 for 1 h at a 0.1 multiplicity of infection (MOI). The medium was subsequently replaced with fresh DMEM or RPMI-1640 containing 3% FBS and the indicated concentration of hyperoside. These cellular supernatants and cells were collected at 24 hours post-infection (hpi) for further qPCR and western blot analysis.
Time-of-addition analysis
To further determine which stage of the PRRSV life cycle was disrupted by hyperoside, MARC-145 cells were seeded into 12-well plates and divided into pretreated, cotreated, and post-treated groups according to the timing of the 120 μM hyperoside treatment relative to PRRSV SD16 (MOI = 0.1) inoculation. Specifically, the pretreated group was incubated with 120 μM hyperoside for 2 h and then infected with PRRSV SD16 (MOI = 0.1). Moreover, the cotreated group was treated with a mixture of hyperoside (120 μM) and PRRSV SD16 (MOI = 0.1) for 1 h. Finally, the post-treated group was infected with PRRSV SD16 (MOI = 0.1) for 1 h, and the medium was then replaced with 3% FBS DMEM containing 120 μM hyperoside. After 24 h, qPCR and western blot assays were performed to examine PRRSV replication in these groups.
To further test whether hyperoside interacts with PRRSV directly, a virucidal activity assay was performed as follows. A mixture of hyperoside (120 μM) and PRRSV SD16 (0.1 and 1 MOI) was incubated together at 37°C for 2 h. Then, MARC-145 cells were treated with this mixture at 37°C for another 1 h. At 24 hpi, the cells and cellular supernatants were collected to assess PRRSV replication via western blot and qPCR assays.
Viral binding, internalization, replication, and release assays
For the binding assay, MARC-145 cells were cultured in 12-well plates and preincubated at 4°C for 1 h. Then, the cells were inoculated with PRRSV SD16 (MOI = 1) and hyperoside (120 μM) or 0.1% DMSO and incubated at 4°C for 2 h to facilitate viral attachment. These cells were then washed three times with prechilled phosphate-buffered saline (PBS) and harvested to extract viral RNA for qPCR quantification.
For the internalization assay, MARC-145 cells were seeded into 12-well plates overnight. These cells were then pretreated with hyperoside (120 μM) for 2 h before infection with PRRSV SD16 (MOI = 1) at 4°C for 1 h to allow viral attachment. The cells were then washed with ice-cold PBS to remove the unbound virus and incubated with 3% FBS DMEM and 120 μM hyperoside or DMSO at 37°C for 1 h. The cells were treated with citrate buffer (pH 3.0) to remove any noninternalized virus particles. Finally, viral RNA was extracted and quantified via qPCR.
For the viral replication assay, MARC-145 cells were cultured in 6-well plates and infected with PRRSV SD16 (MOI = 1) for 6 h. These cells were washed with PBS and then incubated with 3% FBS DMEM containing hyperoside (120 μM) or DMSO at 37°C. The cells were collected at 7, 8, 10, and 16 hpi to quantify PRRSV genome copies via qPCR.
For the viral release assay, MARC-145 cells were cultured in 12-well plates and infected with PRRSV SD16 (MOI = 1) for 24 h. The cells were washed with PBS and incubated with 3% FBS DMEM containing hyperoside (120 μM) or DMSO for 30 or 60 min at 37°C. Finally, the cell supernatants were collected to detect progeny virus particles via qPCR.
Small-interfering RNA assays
The siRNA knockdown assay was performed as described previously. MARC-145 cells were transfected with siRNAs targeting Nrf2 or the siRNA negative control (siNC) for 12 h and then treated with hyperoside (120 μM) for 2 h. Subsequently, the cells were infected with PRRSV SD16 (MOI = 0.5) and harvested at 24 hpi to analyze the protein expression of p62, Nrf2, Keap1, LC3I, LC3II, and PRRSV N via western blotting. MARC-145 cells or PAMs were transfected with siTLR4 or the siRNA negative control (siNC) for 12 h and then treated with hyperoside (120 μM) for 2 h. Subsequently, the cells were infected with PRRSV SD16 (MOI = 0.1) for 24 h. The levels of IL-6, IL-8, IL-1β, and TNF-α mRNA in the cells were analyzed via qPCR, and the production of IL-6, IL-8, IL-1β, and TNF-α in the supernatants from the PAM cultures was analyzed via enzyme-linked immunosorbent assay (ELISA).
ELISA
The supernatants from the PAM cultures were harvested. IL-6, IL-8, IL-1β, and TNF-α production in the supernatants was analyzed via porcine IL-6, IL-8, IL-1β, and TNF-α ELISA kits (Jianglai, Shanghai, China) following the manufacturer’s instructions.
mCherry-GFP-LC3B puncta assay
A mCherry-GFP-LC3B puncta assay was performed to detect the degree of autophagy induced by PRRSV infection, as described in previous studies (
40). Briefly, MARC-145 cells were seeded on coverslips and placed in 12-well plates. Then, these cells were transfected with the mCherry-GFP-LC3 plasmid (Miaoling plasmid, Wuhan, China) for 12 h, treated with different concentrations of hyperoside (60 μM and 120 μM) or 3-MA (30 μM), and infected with PRRSV SD16 (MOI = 0.5) for 24 h. The cells were fixed with 4% paraformaldehyde and stained with DAPI (Invitrogen, USA) for 15 min. Finally, the cells were observed and imaged via an inverted fluorescence microscope (DMi8, Leica, Germany).
Animal experiments
Sixteen 6-week-old PRRSV-negative piglets purchased from Xilingjiao Science and Technology Ltd. (Jinan, China) were randomly divided into four groups (four piglets/group). Group 1 was infected with PRRSV SD16 only (3 × 105 TCID50/piglet), while Group 2 was treated with hyperoside (75 mg/kg) and then challenged with PRRSV SD16 (3 × 105 TCID50/piglet). Group 3 was treated with hyperoside (150 mg/kg) and challenged with PRRSV SD16 (3 × 105 TCID50/piglet). Finally, Group 4 served as the negative control group (mock group). At ?1, 1, 3, 5, and 8 days post-challenge (DPC), piglets from Group 2 and Group 3 received the same oral dose of hyperoside. Moreover, piglets from Group 1 and Group 4 received an equal volume of MEM orally (including 0.1% DMSO). Piglets from each group were housed in separate rooms to prevent cross-infection. Clinical symptoms and rectal temperature were monitored daily.
All the piglets were euthanized, and their lungs were collected for pathological analysis and immunohistochemistry examinations at 28 DPC. Necropsy and pathological analysis were performed as described in our previous study (
36), and pathological scores were calculated. The PRRSV loads in the serum were detected at 4, 7, 10, 14, 21, and 28 DPC, as described previously (
41).
Statistical analysis
The data are presented as the means ± SDs and were analyzed with GraphPad Prism 8.0 software. The intensities of the western blot bands were analyzed via ImageJ 1.8.0 software (National Institutes of Health). Differences among the groups were analyzed via the unpaired Student’s t-test. Statistically significant and very significant differences were defined as those with P < 0.05 and P < 0.01, respectively.
DISCUSSION
PRRSV is an important pathogen that causes serious disease in pigs and hinders the development of the global swine industry. Current vaccines and drugs do not provide adequate protection against this virus, and new antiviral therapies are urgently needed. In the present study, hyperoside was found to exert potent anti-PRRSV effects in susceptible cells (
Fig. 1 and 2) and piglets (
Fig. 9). Furthermore, our mechanistic evaluation revealed that hyperoside alleviates pro-inflammatory responses by inhibiting the TLR4/NF-kB signaling pathway. Additionally, it suppresses PRRSV-induced autophagy via p62/Nrf2/Keap1 axis activation (
Fig. 10).
Viral infection involves multiple phases, namely, binding, internalization, replication, and release (
54). Recent studies have reported that hyperoside exerts antiviral effects at different stages of viral infection. For example, Wang et al. reported that hyperoside inhibits EHV-8 infection by disrupting the adsorption and internalization phases (
17). In contrast, in another study, hyperoside was found to suppress SARS-CoV-2 infection by disrupting viral entry and replication (
55). Furthermore, Wang et al. reported that hyperoside inhibits PEDV infection by preventing the interaction between the N protein and p53 in the replication phase (
10). Notably, the results of our study demonstrated that hyperoside prevents PRRSV infection at the binding, entry, replication, and release stages without causing direct virus inactivation (
Fig. 3).
The inflammatory response represents the host’s defence against viral infections. However, uncontrolled inflammatory responses usually cause severe damage to the host (
56). PRRSV infection triggers the uncontrolled production of pro-inflammatory cytokines (IL-6, IL-8, IL-1β, and TNF-α) to promote self-replication (
57). Interestingly, our study revealed that hyperoside can attenuate the upregulation of IFN-β, IL-6, IL-8, and TNF-α induced by PRRSV infection in susceptible cells (
Fig. 4). NF-κB is a crucial mediator of inflammatory responses (
58), and previous studies have shown that HP-PRRSV can induce pro-inflammatory cytokine production by regulating the TAK-1/NF-κB signaling pathway (
59). Kim et al. reported that hyperoside exerts anti-inflammatory effects by blocking NF-κB activation in mouse peritoneal macrophages (
60). In the present study, we explored the anti-inflammatory mechanisms of hyperoside against PRRSV infection. Our results revealed that hyperoside treatment reduces TLR4, p-NF-κB p65, and p-IκB expression in virus-infected cells (
Fig. 5). Our data suggest that hyperoside can attenuate PRRSV-induced inflammatory storms by inhibiting the TLR4/NF-κB signaling pathway.
The relationship between viral replication and autophagy has been explored extensively. Autophagy can serve as a pro- or antiviral process during virus infection (
61). Diao et al. demonstrated that PRRSV infection induces autophagy via endoplasmic reticulum stress-induced calcium signaling, which promotes viral replication (
40). In the present study, we demonstrated that hyperoside obviously decreased the ratio of LC3II/I expression in PRRSV-infected cells in a dose-dependent manner (
Fig. 6). Additionally, we found that hyperoside can inhibit PRRSV-induced autophagic plaque formation in susceptible cells (
Fig. 7). In addition, our findings revealed that hyperoside has powerful anti-PRRSV activity both
in vitro and
in vivo. However, the present study has several limitations, and further investigations are needed to explore the direct targets of hyperoside.
Conclusions
In conclusion, our results revealed that hyperoside inhibited viral binding, entry, replication, and release during PRRSV infection in a dose-dependent manner. In addition, hyperoside alleviated the inflammatory responses and autophagy caused by PRRSV by suppressing the TLR4/NF-κB pathway and activating the p62/Nrf2/Keap1 axis. Our findings suggest that hyperoside has potential as a promising therapeutic candidate for PRRSV control, warranting further investigations and preclinical studies to evaluate its efficacy and safety.
ACKNOWLEDGMENTS
We thank Shujing Wang from Shandong Agricultural University for providing support in performing the laser confocal experiments in the revised manuscript.
This work was funded by the Shandong Province Pig Industrial Technology System (SDAIT-08-01); Shandong Provincial Natural Science Foundation (ZR2024MC162); the Innovation and Entrepreneurship Program for College Students (CXCY307); the Key Research and Development Project in Shandong Province (2022CXPT010, 2022TZXD0041); and the Taishan Scholars Program.
T.W., L.C., and A.W.: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing—original draft, Writing—review and editing.
Y.X., Y.Z., Q.Z., Q.R., and S.L.: Data curation, Formal analysis, Investigation, Methodology, and Writing—review and editing. Y.L. and J.W.: Writing—review and editing, supervision, and validation.
L.L., Y.L., and J.W.: Conceptualization, Funding acquisition, Validation, Methodology, Project administration, Resources, Software, Supervision, Writing—original draft, Writing—review and editing.
All the authors have read and approved the published version of the manuscript.