key: cord-0074057-ohzgl7bm authors: Huan, Changchao; Zhang, Wei; Xu, Yao; Ni, Bo; Gao, Song title: Antiviral Activity of Plantago asiatica Polysaccharide against Pseudorabies Virus In Vitro date: 2022-01-20 journal: Oxid Med Cell Longev DOI: 10.1155/2022/3570475 sha: 5a7d99ebfa9dd8388c706a4e90cdd117b2c18b8c doc_id: 74057 cord_uid: ohzgl7bm Pseudorabies (PR) is an acute infectious disease of various domestic animals and wild animals caused by pseudorabies virus (PRV). It is mainly characterized by fever, itching, encephalomyelitis, and respiratory and neurological disorders. Plantago asiatica polysaccharide (PLP), extracted from the whole plant of Plantago asiatica L., showed immunomodulatory and antioxidation effects, but the antiviral activity had not been reported. In this study, the inhibitory effect of PLP on PRV infection was studied. Our study first revealed that PLP could inhibit PRV infection in a dose-dependent manner. By adding PLP at different stages of the virus's life cycle, we revealed that PLP could reduce the attachment and penetration of PRV into PK15 cells. The inhibition of PRV attachment was better than inhibition of PRV penetration. However, PLP did not affect PRV replication and inactivation. In addition, PLP decreased the intracellular ROS levels in infected cells significantly, and ROS scavenger NAC decreased PRV infection. Therefore, our study provided preliminary data of anti-PRV activity of PLP, which was established to be a novel anti-PRV infection agent. Pseudorabies (PR) represents the acute infectious disorder in domestic and wild animals resulting from pseudorabies virus (PRV). It is mainly characterized by fever, itching, encephalomyelitis, and respiratory and neurological disorders [1, 2] . It spreads mainly through direct contact or airborne droplets containing the virus [3] . PR first occurred in 1813, and the infected cattle showed extreme itching symptoms, similar to rabies; therefore, it was called "pseudorabies" [4] . PR was transmitted from cattle to pigs after the 1970s [5] . Since 2011, the new PRV variant has led to an epidemic in the pig immunization field in China [6, 7] . Due to the poor protective effect of the classical vaccine, it caused huge losses and resulted in great resistance to the purification of pseudora-bies [8] . PRV was recently isolated from a patient with acute human encephalitis [9] . Therefore, to reduce the economic loss caused by PRV, developing novel antiviral agents for inhibiting PRV infection is of great urgency. Extracts from the Chinese herbal medicines, particularly the corresponding polysaccharides, have shown antiviral activity. Radix isatidis polysaccharide [10] , Platycodon grandiflorus polysaccharides [11] , and kumazasa extract [12] were verified to inhibit PRV infection. The efficacy of such polysaccharides is associated with PRV replication inhibition, the direct killing of PRV, and downregulation of PRV-induced autophagy. These studies offer the foundation to apply polysaccharides in a clinic. Plantago asiatica L., as a Plantaginaceae family member, can be easily cultivated and is widely distributed in eastern Asia [13] . Plantago asiatica L. is used to be a folk medicine since ancient times [14] . As revealed by pharmacological articles, plantain possesses antiulcer, antidiarrheal, antiinflammatory, analgesic, antioxidant, anticancer, and antiviral activities [15, 16] . Phytochemical studies showed that the main chemical components of Plantago asiatica L. are lipids, polysaccharides, iridoid glycosides, alkaloids, flavonoids, and caffeic acid derivatives [13] . Polysaccharides in Plantago asiatica L. (PLP) leaves have several biological activities. PLP displays the effect of immunomodulation through the stimulation of pro-inflammatory cytokine generation [17] . PLP is suggested to promote the production of TNF-α and nitric oxide (NO) via macrophage activation [18] . However, the antiviral activity of PLP has not been reported, and PK15 cells are generally used in in vitro PRV research [19] . Thus, we studied PLP's role in resisting PRV within PK15 cells. Therefore, our work evaluated the antiviral activity of PLP against PRV in vitro and showed that PLP plays an anti-PRV role by mainly inhibiting the attachment and penetration of PRV and decreasing ROS production. Our findings can offer guidance for PLP application, especially in clinical applications as antiviral medicines. We cultivated cells at 37°C and 5% CO 2 conditions and cultivated PK15 cells within DMEM containing 5% fetal bovine serum (FBS), whereas Vero cells within DMEM contained 6% FBS. All the viruses were preserved at -80°C. We isolated PRV XJ5 strain in our laboratory. 2.2. Chemicals, Reagent, and Antibodies. Plantago asiatica polysaccharides (≥98% (uv)) obtained from Yangling Ciyuan Biotechnology Co., Ltd., China was diluted with PBS to 50 mg/mL, followed by preservation at -20°C. The citric acid solution was constituted by citric acid (40 mM), NaCl (135 mM), and KCl (10 mM), and pH was adjusted to 3.0. DAPI was provided by Beyotime Biotechnology (Shanghai, China). In addition, the PRV gB antibody and the PRV-positive serum were prepared and preserved at our laboratory. We obtained a β-actin antibody in Beijing genetically modified Biotechnology Co., Ltd. (Beijing, China), and the goat anti-porcine IgG antibody labeled with FITC was purchased from Sigma-Aldrich (St. Louis, MO). Assay. PK15 cells (5 × 10 5 /well) were laid in a six-well plate and cultivated under 5% CO 2 and 37°C conditions until they grew to a density of 70-80%. After discarding the original culture medium, we rinsed cells thrice using PBS. We cultured PK15 cells within DMEM that contained various concentrations of PLP (100, 200, 400, and 600 μg/mL) for a 1 h period at 37°C. After that, we inoculated PRV XJ5 (multiplicities of infection ðMOIÞ = 0:1) into cells. After infection for 1 h, we discarded virus solution and rinsed cells thrice using PBS. Then, we cultured cells in 2% DMEM containing PLP (100, 200, 400, and 600 μg/ mL) at 37°C. After 24 h postinfection (hpi), we harvested cells to detect viral proteins in cells through WB analysis and collected supernatants for detecting virus titers through TCID 50 . We pretreated PK15 cells for a 1 h period with PLP (600 μg/mL), followed by another 1 h PRV XJ5 (MOI = 0:1) infection. Then, we rinsed cells thrice using PBS, followed by the culture in 2% DMEM that contained PLP (600 μg/ mL). At 2, 4, 8, 12 , and 24 hpi, we measured gB protein expression through WB assay. PK15 cells were pretreated with PLP (400 and 600 μg/mL) and infected with different MOI of PRV XJ5 (MOI = 0:1, 0:5, 1, and 2) for a 1 h period. Then, we rinsed cells thrice using PBS, followed by the culture within 2% DMEM containing PLP (400 and 600 μg/mL). At 24 hpi, we harvested cells for detecting gB protein level by WB assay. Assay. PK15 cells were pretreated with corresponding PLP concentrations (100, 200, 400, and 600 μg/ mL) for a 1 h period at 37°C. After washing thrice using PBS, cells were inoculated with PRV XJ5 (MOI = 0:1) for a 1 h period. Cell supernatants were eliminated, followed by cell rinsing thrice by PBS. Finally, we incubated PK15 cells at 37°C within 2% DMEM. At 24 hpi, we harvested cells for detecting the gB protein level through WB analysis. 2.5. Attachment and Penetration Assay. We rinsed PK15 cells thrice using prechilled PBS, followed by infection using PRV XJ5 (MOI = 0:1) in DMEM containing PLP (100, 200, 400, and 600 μg/mL) for a 1 h period under 4°C without pretreatment. After rinsing cells using cold PBS thrice, cells were cultured with 2% DMEM with PLP (100, 200, 400, and 600 μg/mL) under 37°C. At 1 h later, we discarded cell supernatants. The citric acid solution and PBS removed the uninternalized virus particle, followed by a culture of infected cells within 2% DMEM without PLP. Cells were collected at 24 hpi to detect viral protein levels within cells through the WB assay. Then, we collected cell supernatants for detecting virus titers through TCID 50 . 2.6. Attachment Assay. We rinsed PK15 cells thrice using prechilled PBS, followed by PRV XJ5 (MOI = 0:1) infection with PLP (100, 200, 400, and 600 μg/mL) at 4°C for a 1 h period. After washing thrice using cold PBS, we kept cells with 2% DMEM without PLP for 1 h at 37°C. We harvested cells at 24 hpi, to detect viral protein levels in cells through WB, and we collected cell supernatants for detecting virus titers through TCID 50 . 2.7. Penetration Assay. We infected PK15 cells by PRV XJ5 (MOI = 0:1) for a 1 h period at 4°C with PLP. The cell supernatant was discarded, followed by a culture with 2% DMEM containing PLP (100, 200, 400, and 600 μg/mL) for a 1 h period under 37°C. We eliminated uninternalized virus particles through a citric acid solution and PBS and cultivated those infected cells within 2% DMEM without PLP. Cells and the supernatant were collected at 24 hpi to measure the intracellular viral proteins and virus titer by western blot or TCID 50 . 2.12. Indirect Immunofluorescence Assay (IFA). We fixed cells using the 4% paraformaldehyde (PFA) at 37°C for a 30 min period, followed by 10 min incubation using 0.1% Triton X-100 and overnight sealing using 5% BSA at 4°C. After washing by PBS thrice, we cultured cells using PRVpositive porcine serum for a 1 h period under 37°C and then using FITC-labeled goat anti-porcine IgG antibody for a 30 min period at 37°C. Finally, we stained cells for a 7 min period using DAPI, followed by observation using a fluorescence microscope. 2.13. qRT-PCR. We conducted qRT-PCR according to the previous description [20] . In brief, viral DNA was isolated, followed by assessment of gene levels with specific primers: 2.14. Virus Titer Assays. The virus titer was evaluated by TCID 50 , as performed as the previous description [20] . The results were displayed in the form of mean ± SD and examined using GraphPad Prism software (GraphPad Software, San Diego, CA), and all experiments were repeated independently thrice. One-way ANOVA, as well as Duncan's multiple range test, was utilized to analyze differences between groups. * P < 0:05, * * P < 0:01 stood for significant difference. Thereafter, we infected cells by PRV XJ5 (MOI = 0:1) for a 1 h period with PLP and rinsed them three times using PBS. Afterwards, we cultivated cells with 2% DMEM that contained PLP (100, 200, 400, and 600 μg/mL) at 37°C. At 12 and 24 hpi, the cytopathic effect (CPE) caused by PRV infection was observed using a microscope. Figure 1(a) shows that PLP could reduce CPE in PK15 cells, suggesting that PLP can inhibit PRV infection. The western blot results further confirmed PRV gB protein expression was reduced significantly at PLP concentrations of 400 and 600 μg/mL (Figure 1(b) ). Furthermore, the virus titer in the cell supernatant was evaluated. The results showed that PLP reduced virion production dose dependently (Figure 1(c) ). Additionally, based on IFA results, the number of infected cells decreased with the increase of PLP concentration (Figure 1(d) ). According to the above findings, PLP inhibited PRV infection within PK15 cells. For better investigating PLP's antiviral activity in PRV, we infected PK15 cells by PRV XJ5 (MOI = 0:1) with PLP (Figure 2(a) ). Furthermore, PK15 cells in the presence of PLP (400 and 600 μg/mL) were subjected to PRV XJ5 infection at MOI (0.1, 0.5, 1, and 2). According to WB assay at 24 hpi, PLP inhibited different MOI PRV infections, indicating the highest antiviral activity at the decreased MOI (Figure 2(b) ). To explore the antiviral effect of PLP in various PRV life cycle stages, we pretreated PK15 cells using different concentrations of PLP (100, 200, 400, and 600 μg/mL) followed by 24 h infection by PRV XJ5 (MOI = 0:1) without PLP. As revealed by WB assay, PRV gB protein level was downregulated (Figure 3(a) ), suggesting that pretreatment of cells affected the virus infectivity. For investigating PLP's role in virus attachment and penetration, we infected PK15 cells for a 1 h period by PRV XJ5 (MOI = 0:1) at 4°C that contained different concentrations of PLP (100, 200, 400, and 600 μg/mL). After rinsing by prechilled PBS thrice, we incubated cells by PLP for a 1 h period under 37°C and rinsed thrice by the citric acid solution and PBS for removing those noninternalized virus particles and maintained in 2% DMEM. We harvested cells at 24 hpi for detecting intracellular viral proteins using western blot. The results showed that the protein level of PRV gB reduced, especially with 400 and 600 μg/mL PLP Figure 3(b) ). The supernatant was collected to measure viral titers, which revealed that PRV titer decreased dose dependently (Figure 3(c) ). Besides, according to IFA assay, PLP decreased numbers of infected cells (Figure 3(d) ). These results confirm that PLP inhibited PRV attachment and penetration into PK15 cells. 3.3. PLP Inhibits PRV Attachment to PK15 Cells. To clarify whether PLP could inhibit PRV attachment onto PK15 cells, we infected cells by PRV XJ5 (MOI = 0:1) in DMEM containing PLP (100, 200, 400, and 600 μg/mL) for a 1 h period at 4°C and rinsed them thrice using prechilled PBS. Then, we cultivated cells with 2% DMEM without PLP under 37°C. At 24 hpi, the PRV gB protein expression was measured through WB. As a result, PRV gB protein decreased at PLP concentrations of 400 and 600 μg/mL (Figure 4(a) ). The supernatant was harvested for determining virus titer through TCID 50 , and the results indicated that PRV titers decreased (Figure 4(b) ). According to the IFA assay result, PLP decreased the numbers of infected cells (Figure 4) . These results confirmed that PLP inhibited PRV attachment to PK15 cells. To clarify whether PLP could suppress PRV replication, we incubated PK15 cells by PLP (100, 200, 400, and 600 μg/mL) after 1 h infection by PRV XJ5 (MOI = 0:1) at 37°C. We harvested cells at 4 and 6 hpi to detect PRV gB protein expression levels. The western blot results showed that PRV gB protein expression did not change. In addition, qRT-PCR was carried out to check the change in PRV DNA copy numbers. PLP was found not to affect PRV DNA copy numbers; thus, indicating that PLP did not affect virus replication (Figures 6(a) and 6(b) ). 3.6. Effect of PLP on Virus Inactivation. PRV XJ5 (MOI = 0:1 ) were pretreated by PLP (100, 200, 400, and 600 μg/mL), and the pretreatment virus was inoculated to Vero cells to determine viral titers to clarify whether PLP could directly inactivate the virus. The TCID 50 results showed that the virus titer of the pretreated virus did not change (Figure 7(c) ); thus, confirming that PLP cannot directly inactivate the virus. 3.7. PLP Reduces Intracellular ROS Levels. PK15 cells were treated as described previously, followed by measurement of ROS contents within PK15 cells by DCFH-DA. The results are observed from Figures 7(a) and 7(b) . PLP exposure markedly reduced intracellular ROS levels. Next, whether N-acetylcysteine (NAC) could inhibit PRV infection by reducing ROS production was explored. We treated PK15 cells by NAC (2, 4, 8, or 10 mM) within DMEM for a 1 h period at 37°C. Subsequently, we inoculated PRV (MOI = 0:1) into cells. After removing NAC-virus supernatant at 1 hpi, we rinsed cells thrice by PBS. After cell incubation by the related NAC doses within 2% DMEM, at 24 hpi, PRV gB protein level decreased dose dependently, suggesting that NAC could inhibit PRV infection by reducing ROS production. In the past few years, scientists have been working on vaccines and antiviral medicines; however, PR is still a widespread infectious disease in the swine industry [6] . Since the outbreak of PRV variants in 2011, it induced great economic losses in the swine industry [8, 21] . In addition, the report of human infection with PRV has also attracted global attention [9, 22] . Therefore, the development of new anti-PRV medicines is of great significance. Plant polysaccharides are biological macromolecules widely found in the roots, stems, and leaves of plants. Many studies have reported the antiviral effect of plant polysaccharides [23, 24] . Isatis root polysaccharide can inhibit H3N2 swine influenza virus in vivo and in vitro [25] . Polygonum cillinerve polysaccharide plays antiviral activity via inhibiting transmissible gastroenteritis virus of swine (TGEV) replication, reducing TGEVmediated apoptosis and ROS level [26] . Therefore, plant polysaccharides could be a potential source of antiviral medicines. Recently, researchers mainly focused on polysaccharides in Plantago asiatica L seeds. Evidence showed that polysaccharides in Plantago asiatica L. seeds could alleviate intestinal barrier injury mediated by nonylphenol [27] , resist lipopolysaccharide-mediated liver injury [28] , induce dendritic cell maturation [29, 30] , and promote lipid metabolism and the development of colonic flora [31] . Compared with the seeds, the whole plants have advantages of lower cost and higher utilization rate. Therefore, the polysaccharide in Plantago asiatica L. whole plant was selected to evaluate its antiviral activity in vitro. According to our results, PLP's antiviral activity in PRV in PK15 cells was evaluated. The results showed that PLP decreased the CPE induced by PRV infection and intracellular viral protein expression dose dependently, which confirmed PLP's antiviral activity in PRV. The first step of PRV infection is that virions bind to heparan sulfate proteoglycans in the extracellular matrix and adsorb to the cell surface. Our study revealed that PLP could reduce PRV attachment in a dose-dependent manner, followed by fusion of viral envelope into the cell membrane, making viral capsid penetrate the cytoplasm [1] . Our study confirmed that PLP decreased PRV penetration into PK15 cells. However, the inhibition of PRV attachment was better than inhibition of PRV penetration. In addition, PLP-pretreated cells could reduce the infectivity of PRV. These results suggested that the antiviral mechanism of PLP may be due to the negative charge on the surface of PLP that interacts with cells' surface protein through electrostatic interaction, thus interfering with the virus attachment to the cells [32, 33] . However, PLP did not affect PRV replication and inactivation. ROS accounts for the broad definition of oxidizing compounds in organisms, including hydroxyl radical and hydrogen peroxide [34] . Under normal circumstances, intracellular ROS is at a low level but increases rapidly after the virus invades cells, leading to oxidative stress and cell damage [35] . Our results showed that the intracellular ROS level increased after PRV infection in PK15 cells, and the ROS level decreased significantly after PLP treatment, almost reaching the normal level. Related studies have reported that plant polysaccharides have antioxidant and ROS reduction effects [36, 37] . Ganoderma atrum polysaccharide can decrease the ROS production in immune organs of immunosuppressive animals and reduce the oxidative damage of immune organs [38] . Bletilla striata polysaccharides can reduce angiotensin II-induced oxidative stress and reduce ROS production through NOX4 and TLR2 pathways [39] . In addition, ROS scavenger NAC decreased PRV infection, which confirmed that PLP might reduce ROS to inhibit PRV infection. Our previous studies have shown that the small molecular medicine EGCG can inhibit PRV and PEDV adsorption, entry, and replication [20, 40] . However, most of the small molecule medicines can only act on a single target. Conversely, plant polysaccharides may be more effective in treating or preventing diseases due to their multicomponent and multifunctional characteristics [41] . Plant polysaccharides are safe with immunoregulation, antioxidation, and antiviral effects and exhibit a wide application prospect in preventing or treating diseases [42] . In conclusion, we report the inhibitory effect of PLP on PRV infection within PK15 cells. According to this study, PLP plays an anti-PRV role mainly by inhibiting the attachment and penetration of PRV and decreasing ROS. This study can guide the application of PLP, especially for its clinical application as antiviral medicines. We have already included the available data in our manuscript. The authors have declared no competing interests. Molecular biology of pseudorabies virus: impact on neurovirology and veterinary medicine Comparative pathology of pseudorabies in different naturally and experimentally infected species-a review Pseudorabies virus infections in pigs Aujeszky's disease (pseudorabies) virus: the virus and molecular pathogenesis-state of the art Control of swine pseudorabies in China: opportunities and limitations Current status and challenge of pseudorabies virus infection in China Pathogenicity and genomic characterization of a pseudorabies virus variant isolated from Bartha-K61-vaccinated swine population in China Pseudorabies virus variant in Bartha-K61-vaccinated pigs A novel human acute encephalitis caused by pseudorabies virus variant strain Antiviral activities of Radix isatidis polysaccharide against pseudorabies virus in swine testicle cells Platycodon grandiflorus polysaccharides inhibit pseudorabies virus replication via downregulating virus-induced autophagy Anti pseudorabies virus activity of kumazasa extract The traditional uses, chemical constituents and biological activities of _Plantago major_ L. A review The effect of antioxidant and whitening action on Plantago asiatica L. leaf ethanol extract for health care Chemical constituents and medical benefits of Plantago major Plantago major in traditional Persian medicine and modern phytotherapy: a narrative review Molecular properties and immunomodulatory activities of a water-soluble heteropolysaccharide isolated fromPlantago asiaticaL. leaves Polysaccharides extracted from the leaves of _Plantago palmata_ Hook.f. induce nitric oxide and tumor necrosis factor-α production by interferon-γ-activated macrophages Growth characteristics and complete genomic sequence analysis of a novel pseudorabies virus in China Epigallocatechin-3-gallate inhibits the life cycle of pseudorabies virus in vitro and protects mice against fatal infection Emergence of a pseudorabies virus variant with increased virulence to piglets Human endophthalmitis caused by pseudorabies virus infection The effect of Astragalus polysaccharide on the Epstein-Barr virus lytic cycle Anti-herpes activity of polysaccharide fractions from Stevia rebaudiana leaves Effects of Isatis root polysaccharide in mice infected with H3N2 swine influenza virus The inhibitory effect Polygonum cillinerve polysaccharide on transmissible gastroenteritis virus of swine Polysaccharide from the seeds of Plantago asiatica L. alleviates nonylphenol induced intestinal barrier injury by regulating tight junctions in human Caco-2 cell line Polysaccharide from the seeds ofPlantago asiaticaL. protect Against Lipopolysaccharide-Induced Liver Injury Polysaccharide isolated from seeds of Plantago asiatica L. induces maturation of dendritic cells through MAPK and NF-kappaB pathway A novel polysaccharide from the seeds of Plantago asiatica L. induces dendritic cells maturation through toll-like receptor 4 Polysaccharide from seeds of Plantago asiatica L. affects lipid metabolism and colon microbiota of mouse The antiviral activity of polysaccharides and their derivatives Recent advances in antiviral activities and potential mechanisms of sulfated polysaccharides ROS and diseases: role in metabolism and energy supply Apoptosis induction by pseudorabies virus via oxidative stress and subsequent DNA damage signaling Ganoderma lucidum polysaccharide inhibits UVB-induced melanogenesis by antagonizing 12 Oxidative Medicine and Cellular Longevity cAMP/PKA and ROS/MAPK signaling pathways Astragalus polysaccharide attenuates cisplatin-induced acute kidney injury by suppressing oxidative damage and mitochondrial dysfunction Ganoderma atrum polysaccharide ameliorates ROS generation and apoptosis in spleen and thymus of immunosuppressed mice Bletilla striata polysaccharide inhibits angiotensin II-induced ROS and inflammation via NOX4 and TLR2 pathways Epigallocatechin-3-gallate, the main polyphenol in green tea The anti-oxidant and antitumor properties of plant polysaccharides Application prospect of polysaccharides in the development of anti-novel coronavirus drugs and vaccines The present work was supported by the Natural Science