Next Article in Journal
Wheat Susceptibility Genes TaCAMTA2 and TaCAMTA3 Negatively Regulate Post-Penetration Resistance against Blumeria graminis forma specialis tritici
Previous Article in Journal
Effects of Exercise Preconditioning on Doxorubicin-Induced Liver and Kidney Toxicity in Male and Female Rats
Previous Article in Special Issue
Analysis of the Electron Density of a Water Molecule Encapsulated by Two Cholic Acid Residues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploring the Inhibitory Effect of AgBiS2 Nanoparticles on Influenza Viruses

1
Institute of Bismuth and Rhenium Science, School Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
2
Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
3
CAS Key Laboratory of Molecular Virology & Immunology, Institutional Center for Shared Technologies and Facilities, Pathogen Discovery and Big Data Platform, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
4
School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
5
Institutional Center for Shared Technologies and Facilities of Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(12), 10223; https://doi.org/10.3390/ijms241210223
Submission received: 25 April 2023 / Revised: 2 June 2023 / Accepted: 12 June 2023 / Published: 16 June 2023
(This article belongs to the Special Issue Nano-Materials and Methods 4.0)

Abstract

:
Influenza viruses are respiratory pathogens that are major threats to human health. Due to the emergence of drug-resistant strains, the use of traditional anti-influenza drugs has been hindered. Therefore, the development of new antiviral drugs is critical. In this article, AgBiS2 nanoparticles were synthesized at room temperature, using the bimetallic properties of the material itself to explore its inhibitory effect on the influenza virus. By comparing the synthesized Bi2S3 and Ag2S nanoparticles, it is found that after adding the silver element, the synthesized AgBiS2 nanoparticles have a significantly better inhibitory effect on influenza virus infection than Bi2S3 and Ag2S nanoparticles. Recent studies have shown that the inhibitory effect of AgBiS2 nanoparticles on the influenza virus mainly occurs in the stages of influenza virus-cell internalization and intracellular replication. In addition, it is found that AgBiS2 nanoparticles also have prominent antiviral properties against α and β coronaviruses, indicating that AgBiS2 nanoparticles have significant potential in inhibiting viral activity.

1. Introduction

The influenza virus is a kind of respiratory pathogen. After a mild infection, it can cause symptoms, such as runny nose, fever, and cough. In severe cases, it can cause pneumonia, which is one of the major disease endangering the life and safety of the patient. The US Food and Drug Administration (FDA) has only approved two types of drugs to treat influenza viruses, namely M2 protein ion channel blockers and neuraminidase inhibitors [1]. With the long-term use of drugs, drug-resistant strains have emerged which have restricted the use of these two types of anti-influenza virus drugs. This has become a public health problem worldwide. On the other hand, vaccines for influenza viruses often fail as the virus mutates. Therefore, it is necessary to find new anti-influenza virus drugs or broad-spectrum antiviral drugs with different antiviral mechanisms to replace the traditional drugs.
In recent years, nanomaterials have been rapidly developed and are widely used in biotherapy. In addition, with the deepening of research, it has been found that metal nanomaterials have antibacterial [2], anti-tumor [3], anti-virus [4], and anti-parasitic properties [5]. Studies have found that Ag and Au nanoparticles have prominent antiviral properties. Their action occurs before the virus invades the cell and during the process of crossing the cell membrane. However, some things could be improved in the antiviral materials reported at this stage. First of all, the inhibitory effect of nanomaterials is mainly to block virus adsorption and entry into cells, and there is no apparent inhibitory effect in the late stage of virus replication. Secondly, the application of single metal has certain limitations. Therefore, multi-metal nanomaterials are used in antiviral therapy to enrich the characteristics of nanomaterials, improve their antiviral performance, and show excellent antiviral capabilities [6,7].
Researchers have found that bismuth compounds have good photothermal conversion ability in the near-infrared region in tumor treatment, so they can be used as a photothermal treatment of tumor reagents and can achieve ideal therapeutic effects. Regarding antibacterial properties, studies have found that the bismuth ions in metallothionein in organisms can interact strongly with cysteine, providing hope for applying bismuth compounds to inhibit bacteria and viruses [8]. At this stage, bismuth coordination compounds, such as bismuth subsalicylate and ranitidine bismuth citrate, can form a protective film in the stomach, prevent gastric acid and protease from harming the stomach, and inhibit Helicobacter pylori. It is widely used to treat gastric ulcers [9,10]. In addition, research has shown that ranitidine bismuth citrate can play an essential inhibitory role in the late stage of the severe acute respiratory syndrome (SARS) virus replication cycle through the coordination of bismuth ions [11]. With the outbreak of the coronavirus disease 2019 (COVID-19), many drugs have been used to restrain or kill the new coronavirus. Studies have shown that bismuth salt inhibits the NTPase and RNA helicase of the new coronavirus type 2 [12]. Moreover, studies have found that ranitidine bismuth citrate, used to treat stomach diseases, can inhibit the replication of new coronavirus type 2 and reduce the symptoms of pneumonia in mice [13].
In addition, Ag also plays a vital role in the field of anti-virus. Park et al. synthesized a kind of magnetic hybrid colloid loaded with Ag nanoparticles of different sizes. Using the interaction between Ag and biological macromolecules, Park explored the inhibitory effect of those nanoparticles on bacteriophages, noroviruses, and adenoviruses. The results show that Ag nanoparticles are combined with sulfhydryl-containing proteins on the virus’s surface, thereby destroying the virus envelope and killing the virus. Therefore, Ag is widely used to prepare antibacterial and antiviral materials due to its good bactericidal and antiviral properties [14].
We synthesized Bi-based nanomaterials and explored their antiviral potential. Their inhibitory effect on influenza virus infection at the cellular level was tested. Nanomaterials were added at different stages of virus-infected cells, and the inhibition effect was evaluated via TCID50, fluorescent quantitative PCR (qRT-PCR) and indirect immunofluorescence methods. Furthermore, the antiviral effect of bimetallic nanomaterial AgBiS2 was intensively investigated. Our research may provide simple, green, efficient, and low-cost antiviral nanomaterials for application.

2. Results

2.1. Material Characterization

Three kinds of nanoparticles (AgBiS2, Bi2S3, and Ag2S) are rapidly synthesized at room temperature, and the synthesized nanoparticles are tested and characterized using TEM and XRD (Figure 1). As shown in Figure 1a, the experimentally synthesized AgBiS2 material is irregularly granular and crystalline, with an average particle size of 18.6 ± 0.3 nm, uniform particle size, and good dispersion. The XRD test of the experimentally synthesized AgBiS2 shows that the XRD diffraction peaks of AgBiS2 nanoparticles are consistent with the standard card JCPDF 89-2045 (Figure 1d). These data indicate the successful synthesis of AgBiS2 nanoparticles. As shown in Figure 1b, the shape of Bi2S3 nanoparticles is similar to that of mulberries, with irregular shapes and a particle size of about 40 nm. In addition, XRD analysis shows that the diffraction peaks of the experimentally synthesized Bi2S3 nanoparticles are consistent with the standard card JCPDF 75-1036 (Figure 1e). As shown in Figure 1c, the experimentally synthesized Ag2S nanoparticle is spherical with an average particle size of 64 nm. TEM shows that part of Ag2S is connected with multiple morphologies because PVP wraps the outer surface. Additionally, the comparison of the Ag2S standard card (JCPDF 89-3840) shows that the XRD diffraction peaks of the test group match the standard card (Figure 1f). In summary, the synthesis of experimental nanomaterials is successful.
In order to verify the successful synthesis of AgBiS2 nanoparticles, the synthesized material was subjected to high-angle circular dark scene scanning (mapping). It can be observed from Figure 2 that the distribution of the three elements of Bi, S, and Ag in nanoparticles further demonstrates the successful synthesis of AgBiS2 nanoparticles.
After synthesizing the three kinds of nanomaterials, their chemical properties were tested. The analysis shows (Figure 3a) that the hydrodynamic diameters of AgBiS2, Bi2S3, and Ag2S are 43.3 nm, 52.1 nm, and 76.3 nm, respectively, based on the hydrated particle size (DLS).
As shown in Figure 3b, the zeta potentials of AgBiS2, Bi2S3, and Ag2S in an aqueous solution are −12.56 mV, −8.03 mV, and −20.5 mV, respectively. The zeta potential of AgBiS2 is between Bi2S3 and Ag2S. With the addition of Ag, the zeta potential value decreased from −20.5 mV to −12.56 mV. The infrared analysis (IR) of the synthesized nanoparticles and PVP, as shown in Figure 3c, highlights that the stretching vibration peak of the amino group (N-H) of PVP was around 3400 cm−1, and that the stretching vibration peak of the carbonyl group (C=O) was at 1685 cm−1. The alkyl group (C-H) was stretched and flexed by the nanoparticles at 2871 cm−1, the alkyl group (C-H) at 3027 cm−1 was the stretching vibration peak, and at 896 cm−1 the alkyl group (C-H) was the rocking vibration peak [15,16]. Thermogravimetric analysis (TG) shows that the weight of Bi2S3 begins to decrease at 200–300 °C (Figure 3d). This stage indicates that the loss of unstable oxygen photo energy groups on the material leads to a decrease in the weight of the material. The decrease in weight at 300–350 °C is attributed to the carbonization of the PVP material wrapped in the outer layer, which separates the PVP wrapped in the outer layer from the Bi2S3 inside. The decrease in weight at 450–550 °C is attributed to the collapse of the material skeleton. For AgBiS2, the 3.46% weight loss at 300–350 °C is lower than that of Bi2S3 at 5.13%. This indicates that in the synthesized materials, the amount of PVP wrapped on the outer surface of Bi2S3 is higher than that of AgBiS2.

2.2. Cytotoxicity Test

Good biological safety is the prerequisite for using nanoparticle in biological therapy. The biocompatibility of synthetic nanoparticle is evaluated by studying the toxicity of three nanoparticles to MDCK cells. As shown in Figure 4, the MTT method detects the cell viability of the three nanoparticles incubating with cells for 24 and 48 h. It is found that when the AgBiS2 concentration reaches 150 μg/mL, the survival rate of MDCK cells after 48 h of co-culture is still higher than 90%, indicating that AgBiS2 has lower cytotoxicity than Ag2S and Bi2S3. However, when Ag2S and Bi2S3 were co-cultured with cells for 48 h, the cell survival rate was higher than 90%, while the concentration was lower than 100 μg/mL. The above results show that the biological safety of AgBiS2 is significantly higher than that of Ag2S and Bi2S3, and that further biological experiments should be carried out within the biological safety concentration of each group of materials.

2.3. Antiviral Effect Evaluation Experiment

2.3.1. Direct Inactivating Group

Firstly, the direct killing of the materials against viruses was tested. Different concentrations of nanoparticles and viruses were incubated at room temperature for 1 h. The incubated virus solution was used to infect cells, and then the relative content of the virus was tested via RT-qPCR after 48 h of co-culture [17]. The results are shown in Figure 5. Although AgBiS2, Ag2S, and Bi2S3 nanoparticles have weak infection inhibition abilities against the influenza virus, the antiviral effects of these three types are not obvious. The infection inhibition rate against the virus was about 20%. It shows that the three kinds of nanoparticles cannot kill the virus through interaction with the virus.

2.3.2. Co-Cultivation Inhibition Group

In order to verify whether nanoparticles interfere with the process of virus adsorption to cells, nanoparticles and influenza viruses were incubated with the cells for an hour at the same time. After washing off unabsorbed influenza viruses and nanoparticles, the RT-qPCR tests were performed after 48 h of incubation [18]. As shown in Figure 6, the virus suppression effect of AgBiS2 and Ag2S is higher than the Bi2S3 group, and the best suppression effect of the Ag2S group is 71.45%. Among them, Ag2S shows a good antiviral effect at a concentration of 10 μg/mL. The inhibition rate of the influenza virus reached 47.69%, which is significantly higher than 40.4% in the AgBiS2 group and 14.79% in the Bi2S3 group. With the increase in the nanoparticles’ concentration, the viral inhibitory effect is improved and tends to be flat. The above results show that Ag2S exhibits the best influenza virus inhibitory effect among the three nanoparticles when the nanoparticles and influenza virus are added to the cell together. The antiviral effect of AgBiS2 is the second best. However, Bi2S3 shows very weak antiviral ability. By comparing the three kinds of nanoparticles, it is found that in the presence of Ag ions, the process of virus adsorption to cells will be significantly interfered, thereby reducing the amount of influenza virus adsorbed to cells and achieving the effect of inhibiting influenza virus infection.

2.3.3. Nanoparticle Preculture Inhibition Group

The preliminary exploration shows that the three kinds of nanoparticles can play a blocking role in the process of virus adsorption into cells. To further determine the antiviral effects of the three nanoparticles in the cell, pre-cultivating the nanoparticles at different concentrations with the cells was carried out for 3 h before incubating with the viruses. After the virus and cells were co-cultured for 1 h, the unabsorbed viruses were washed away. After 48 h of culture, the inhibitory effect of different materials on the influenza virus was determined via RT-qPCR (Figure 7). When the nanoparticles entered the cells earlier than the viruses, the degree of intervention of the nanoparticles on the influenza virus replication inside the cells was examined. As shown in Figure 7a, AgBiS2 exhibits a significant inhibitory effect on influenza viruses at low concentrations. As the concentration of AgBiS2 increases, the inhibition rate reaches 78.78%. The maximum inhibition rate is significantly higher than 64.24% in the Ag2S group and 48.53% in the Bi2S3 group (Figure 7b,c). It indicates that AgBiS2 can interfere with the intracellular replication process of the virus. The reason is that Bi3+ and Ag+ have an affinity for the biological macromolecular protein of viruses and can bind to the biological protein through coordination or chemical reaction so that the protein molecules can be changed in conformation and lose the biological activity [8,19]. Among them, the virus suppression effect of the bimetallic AgBiS2 nanoparticles was significantly better than that of the single metal Ag2S and Bi2S3 nanoparticles.

2.3.4. Virus Post-Infection Inhibition Group

To explore the contribution of nanoparticles’ interference to the transmembrane process of the influenza virus, after the virus was adsorbed on the cell surface, the nanoparticles were added and cultured for 48 h. RT-qPCR shows that adding AgBiS2, Ag2S, and Bi2S3 enhances the inhibitory effect of the influenza virus (Figure 8). The comparison of the inhibitory effects of the three nanoparticles revealed that the maximum inhibitory rate of AgBiS2 on the influenza virus was 91.72%, which is significantly higher than the inhibitory effects of the Ag2S and Bi2S3 groups. In this virus post-infection group, nanoparticles participate in the virus-cell internalization process in the early stage and the virus intracellular replication process in the later stage. Referring to the results of the material preculture group, AgBiS2 and Ag2S can interfere with the replication stage of the influenza virus and suppress virus propagation. In the virus preculture group, nanoparticles affect both the transmembrane and virus replication processes in cells. Both AgBiS2 and Ag2S shows the inhibiting effect on the virus. However, the inhibiting effect of AgBiS2 is higher than that of Ag2S, which indicates that AgBiS2 plays an inhibitory role in the process of virus transmembrane due to the introduction of Bi3+. It can also be mutually confirmed by comparing the virus inhibition effects of Bi2S3 between the virus post-infection inhibition group and the nanoparticle preculture inhibition group.

2.4. TCID50 Test for Virus Activity

After treatment in each experimental group, the virus-containing supernatant was collected, and the TCID50 method was used to detect the virus titer and verify whether the virus in the supernatant was living. After the influenza virus was cultured for six days, the TCID50 of the influenza virus was measured via hemagglutination assay.
For AgBiS2 nanoparticles, as shown in Figure 9, by comparing the four different inhibition pathways, it was found that after AgBiS2 nanoparticles treatment, the influenza virus titer of the group (d) decreases most significantly with 1.58 lg (TCID50), followed by 1.1 lg (TCID50) of the group (c). It indicates that the main inhibitory effect of AgBiS2 nanoparticles on the influenza virus occurs during the cellular internalization and intracellular replication stages. These data further confirm the results of RT-qPCR detection.
For Ag2S nanoparticles, the results of the RT-qPCR test show that the inhibitory effect of Ag2S on the influenza virus is lower than that of AgBiS2. TCID50 is used to determine the live virus in the virus-containing supernatant. It is found that the Ag2S-group (c) has the best inhibitory effect, and the titer of the influenza virus decreases by 1.14 lg (TCID50) (Figure 10), but it is lower than the inhibitory effect of the AgBiS2-group (d) (Figure 9). It indicates that the main inhibitory stage of Ag2S nanoparticles against influenza viruses occurs in the virus’s intracellular replication process.
For Bi2S3 nanoparticles, detection of the influenza virus titer of each experimental group shows that the virus titer of the group (d) decreases. It shows an inhibitory effect on the influenza virus at a lower concentration. The inhibitory effect is low for group (a) and group (b) (Figure 11). The above conclusions are consistent with the results of RT-qPCR.

2.5. Indirect Immunofluorescence Experiment

The viruses mainly invade cells by the fusion of virus surface proteins and cell surface receptors. After the virus is adsorbed on the cell surface, part of the virus antigen protein will be exposed to the cell’s outer layer. As a standard virologic technique, the indirect immunofluorescence assay (IFA) demonstrates the influenza viral antigens expressed in infected cells. The virus antigen can be detected to indirectly determine the degree of infection of the virus to the cell. At this time, the indirect immunofluorescence method can determine the number of virus-infected cells [19,20,21].
Indirect immunofluorescence has been extensively used as a confirmatory assay in viral research. Here, IFA was performed to study the inhibitory impact of nanoparticles on the expression of influenza virus HA antigen. After pre-cultivating the virus and cells for 1 h, AgBiS2 solution at different concentrations (0, 50, and 150 μg/mL) was added, and the cells were fixed on the glass bottom 96-well plate after 24 h. As shown in Figure 12, using a rotating disc confocal microscope it is observed that when the concentration of AgBiS2 nanoparticles is 0 μg/mL; the fluorescence intensity of the virus-positive control group was higher. When the concentration of AgBiS2 nanoparticles is increased to 50 μg/mL, it is found that the HA intensity reduced significantly compared with the positive control group, indicating that AgBiS2 inhibited the process of virus infection. As the concentration of AgBiS2 increased to 150 μg/mL, the fluorescence intensity of HA protein further decreased compared with the 50 μg/mL group. It indicates that AgBiS2 nanoparticles have an inhibitory effect on influenza virus-infected cells, and as the concentration of the added nanomaterial increases, the inhibitory effect increases.

2.6. Inhibitory Effect on Influenza A Virus, α and β Coronaviruses

To verify the broad-spectrum antiviral properties of AgBiS2 nanomaterials, we explored the antiviral effect of AgBiS2 nanomaterials on influenza virus A, α and β coronaviruses. The virus post-infection inhibition group approach was applied. After culturing the AgBiS2 nanomaterials with the infected cells for 48 h, it is observed from Figure 13a that the inhibitory effect on influenza A is significantly increased with the increased concentrations of AgBiS2 nanomaterials. The highest inhibition rate at 100 μg/mL was 90.25%. In addition, we verified the infection-inhibitory effect of AgBiS2 nanomaterials on β coronavirus OC43 and α coronavirus 229E. Figure 13b,c show that the AgBiS2 nanomaterials also have a significant inhibitory effect on coronavirus. As the concentration of AgBiS2 nanomaterials increased, the inhibition effect was the highest at 100 μg/mL, 88.25% and 86.06% for β coronavirus OC43 and α coronavirus 229E, respectively. It indicates that AgBiS2 nanomaterials have broad-spectrum and excellent antiviral properties.

3. Discussion

The research on nanomaterials has become one of the emerging hot spots, greatly expanding its application [22,23]. However, the application of metal nanomaterials was limited due to their poor biological safety for a long time [24]. As the preparation of nanomaterials is improved by adjusting the proportion of raw materials and exploring the appropriate reaction conditions, nanomaterials with small sizes, high specific surface area, adjustable particle size and flexible surface functionalization are developed [25]. Moreover, with the development of metal nanomaterials in anti-tumor research, metal nanomaterials have shown extensive application prospects in the biological field [3,26]. Furthermore, with the recent outbreak of novel coronavirus, researchers are paying attention to treating viral infectious diseases with metal-based nanomaterials [12,13,20].
In this study, we screened the bismuth-based nanomaterials with anti-virus activity, especially the inhibitory effect on the influenza B virus, and explored the possible inhibitory stages. The virus replication was inhibited by PVP-coated AgBiS2 nanomaterials, which is quite similar to Hongze Sun’s lab results, wherein the suppressive effect on SARS-CoV-2 by metallodrug ranitidine bismuth citrate was demonstrated [13]. The three inhibition pathways of metal nanomaterials as antiviral therapeutic materials against viral infections, including direct inactivation [14,21,27,28], inhibition of virus adsorption and entry [29,30,31], and intracellular virus suppression [18,19], are very common [32]. However, further study on the mechanism of AgBiS2′s antiviral effect will identify the related molecules and pathways.
Pure bismuth and unmodified bismuth compound, or their nano crystallized materials, have poor solubility, so we used the PVP-coated AgBiS2 with good biocompatibility which enhanced the nanomaterial’s solubility and its internalization into cells [33,34]. Jiangfeng Du’s work shows that poly(vinylpyrrollidone)- and selenocysteine-modified Bi2 Se3 nanoparticles (PVP-Bi2 Se3@Sec NPs) are in vivo, in vitro low toxicity and biodegradable with dramatic biological effect [33]. Similarly, it is indicated that PEGylation effectively reduces cytotoxicity and increases zinc oxide nanoparticles’ antiviral activity against herpes simplex virus type 1 [34]. Meanwhile, a kind of iron oxide nanoparticles (IO-NPs) developed to fight against the H1N1 influenza A virus was evaluated via MTT and TCID50 and proven to have excellent performance [29]. In addition, the functional modification of nanomaterials was carried out based on the characteristics of virus receptors on the cell surface [25,35]. The strategy proved very effective.
We propose that PVP-AgBiS2 nanomaterials internalized into cells affect the pivotal enzymes related to virus replication. The enzyme motif analysis may help us speculate the interaction model between bismuth and enzymes. A functional study will be performed to elucidate the mechanism of bismuth’s effect on enzymes in viral replication. Hopefully, the in-depth understanding will helpfully reveal the target of bismuth during its inhibition of virus propagation.

4. Materials and Methods

4.1. Synthesis of Nanoparticles

4.1.1. Synthesis of Bi2S3 Nanoparticles

We dissolved 121 mg of Bi(NO3)3·5H2O in 20 mL ethylene glycol, added 300 mg of PVP-k30, and stirred it with ultrasonic until dissolved. In addition, we weighed 20 mg of thiourea, added it to 3 mL of ethylene glycol and 2 mL of ethanol, and dissolved it by ultrasound. Then, we added dropwise the thiourea solution to the Bi(NO3)3 solution, stirred rapidly for 15 min, centrifuged and washed with ethanol and ultrapure water several times, and then dispersed in PBS (pH = 7.4) for later use.

4.1.2. Synthesis of Ag2S Nanoparticles

We weighed 40 mg of AgNO3, dissolved it in 20 mL of ethylene glycol, stirred it to dissolve properly, and then added 300 mg of PVP-k30 to dissolve ultrasonically. Another 20 mg of thiourea was dissolved in 3 mL of ethylene glycol and 2 mL of ethanol ultrasonically. Then, this mixture was added to the AgNO3 solution and stirred rapidly for 15 min, and the centrifugal washing step was the same as the synthesis of bismuth sulfide above.

4.1.3. Synthesis of AgBiS2 Nanoparticles

We took 61 mg of Bi(NO3)3·5H2O and 21 mg of AgNO3 to dissolve them in 20 mL of ethylene glycol, and then added 300 mg of PVP-k30 through ultrasonic blending. Then, we added 20 mg of thiourea, and the other operations were the same as the synthesis of bismuth sulfide above. Bi(NO3)3·5H2O, PVP-k30, AgNO3, thiourea, ethanol, and ethylene glyco were purchased from Sinopharm Group Chemical reagent Co., LTD., Shanghai, China.

4.2. Characterization of Nanoparticles

We observed the morphology and structure of the synthesized nanoparticles through transmission electron microscopy (TEM, HT7800; Hitachi High-Tech, JPN, Tokyo, Japan) and obtained the energy spectrum data of the material through energy dispersion spectroscopy (EDS, Ultra55; Zeiss, GER, Oberkochen, Germany); Use Zeta potential measuring instrument (Nano-zs90; Malvern, UK) to determine the particle size distribution (DLS) and zeta potential of nanoparticles.

4.3. Cultivation of Cells and Viruses

Madin–Darby canine kidney (MDCK) cells (ATCC, CCL-34), an immortal human hepatic cell line HuH-7 (JRCB0403) and RD cells (ATCC, CCL-136), preserved by the Institut Pasteur of Shanghai, Chinese Academy of Sciences, were used as host cells for influenza viruses, coronavirus 229E and OC43, respectively. The cells were cultured in EMEM (ATCC, 302003) supplemented with 10% fetal bovine serum (FBS, Gibco™ 10099141C, Thermo Fisher Scientific Inc., Waltham, MA) and 1% penicillin-streptomycin (PS) in an incubator with 5% CO2 at 37 °C. Influenza virus B/Mass/3/66 (ATCC, VR-523), influenza A Virus A/WS/33 (ATCC, VR-825), human coronavirus 229E (ATCC, VR-740) and OC43 (ATCC, VR-1558) were deposited by the Institut Pasteur of Shanghai, Chinese Academy of Sciences. The virus titer was determined using the 50% cell culture infectious dose (TCID50) endpoint dilution assay, and calculated according to the Reed–Muench formula [36].

4.4. Cytotoxicity Test

The biological cytotoxicity of the prepared nanoparticles was evaluated via the MTT assay. First, the MDCK cells were seeded in a 96-well flat-bottom plate at a density of 104 cells/well, and were next cultured overnight in a 37 °C, 5% CO2 incubator. Then, the cells grew about 80–90% of the density, aspirated the culture medium in the wells, and then we washed the plates twice with PBS and added 100 μL of serum-free medium to each well of nanoparticles at different concentrations (0, 10, 20, 50, 100, 150, and 200 μg/mL) with a blank control, and applied six replicate wells for each concentration, and placed them in a 37 °C, 5% CO2 incubator. After 24 or 48 h, we aspirated and discarded the cell supernatant, washed the plates twice with PBS, and added 100 μL of new serum-free medium. Then, we added 25 μL of MTT (5 μg/mL), put the plates in the incubator for 4 h, added 100 μL of 10% SDS extraction buffer, and measured the absorbance at 570 nm after overnight incubation.
The following formula was used to calculate the nanomaterial’s cytotoxicity (percent coefficient of variation, CV%). CV% = O D s O D m O D n O D m × 100 % . Among them, ODs is the absorbance of the experimental group, ODn is the absorbance of the control group, and ODm is the absorbance of the blank group.

4.5. TCID50 Assay for Determination of Virus Titer

MDCK/HuH-7/RD cells were cultured overnight in 96-well flat-bottom plates at 2×104 cells/well. Then, we discarded the supernatant, washed the plates twice with PBS, and added a series of virus dilutions in 100 μL of virus infection medium (from 10 to 106 dilutions), ten replicates for each dilution, and two 96-well plates in parallel. Then, we placed the 96-well flat-bottom plates in a 35 °C (for influenza viruses) and 37 °C (for coronaviruses), 5% CO2 incubator for 6 days. After that, we transferred 50 μL influenza virus culture supernatant to each well from the 96-well flat-bottom plates to new U-shaped 96-well plates, gently mixed with 50 μL of 1% chicken red blood cells and then recorded the number of non-agglutinate wells after standing for about 30 min. Meanwhile, cytopathic effects (CPE) of coronaviruses were measured using crystal violet staining. The TCID50 was calculated according to the Reed–Muench formula, and a titer of 100 TCID50/mL was used for in vitro cell experiments.

4.6. Virus Inhibition Test

4.6.1. Direct Inactivating Experiment

MDCK cells were cultured overnight in 96-well plates at a density of 2 × 104 cells/well. We used a serum-free medium to prepare the virus with a final concentration of 100 TCID50/mL and materials of different concentrations (0, 10, 20, 50, 100, and 150 μg/mL). Then, we mixed the solution, and incubated at room temperature for one hour. Subsequently, we discarded the supernatant, washed the plates twice with PBS, added 100 μL/well of the viral material mixture and incubated the 96-well plates at 35 °C for 1 h. Then, we washed the plates twice with PBS, added 100 μL/well of viral infection base and incubated at 35 °C for 48 h. Finally, the supernatant was collected from each well and the viral RNA extraction from the supernatant was carried out for RT-qPCR and TCID50 detection [18].

4.6.2. Co-Cultivation Inhibition Experiment

MDCK cells were cultured overnight in 96-well plates at a density of 2 × 104 cells/well. Then, we used a serum-free medium to prepare 200 TCID50/mL virus and nanomaterial dilutions of different concentrations. We discarded the cell supernatant, washed the plate twice with PBS, and added the final concentration of 100 TCID50/mL virus mixed with 0, 10, 20, 50, 100, and 150 μg/mL of material diluent. We placed the 96-well plate at 35 °C for 1 h, then washed the plates twice with PBS, added 100 μL/well of viral infection medium and incubated at 35 °C for 48 h. Finally, we collected the supernatant for each well and extracted the viral RNA from the supernatant for RT-qPCR and TCID50 detection.

4.6.3. Nanoparticle Preculture Inhibition Experiment

MDCK cells were cultured overnight in 96-well plates at a density of 2 × 104 cells/well. We used a serum-free medium to prepare material dilutions with 0, 10, 20, 50, 100, and 150 μg/mL concentrations. Then, we discarded the cell supernatant, washed the plate twice with PBS, added 100 μL/well of material diluent and incubated it at 37 °C for 3 h. Next, we washed the plates twice with PBS, added 100 μL/well of 100 TCID50/mL virus solution and incubated at 35 °C for 1 h. Again, we washed the plates twice with PBS, added 100 μL/well of viral infection medium and incubated at 35 °C for 48 h. Finally, we collected the supernatant for each well and extracted the viral RNA from the supernatant for RT-qPCR and TCID50 detection.

4.6.4. Virus Post-Infection Inhibition Experiment

MDCK cells were cultured overnight in 96-well plates at a density of 2 × 104 cells/well. We discarded the cell supernatant, washed the plate twice with PBS, added 100 μL/well of 100 TCID50/mL virus solution prepared with a serum-free medium, and incubated at 35 °C for 1 h. Then, we discarded the cell supernatant, washed the plates twice with PBS and add 100 μL/well of material dilutions prepared with viral infections at concentrations of 0, 10, 20, 50, 100, and 150 μg/mL, and incubated at 35 °C for 48 h. Then, we collected the supernatant for each well and extracted the viral RNA from the supernatant for RT-qPCR and TCID50 detection, respectively.

4.7. Real-Time Fluorescent Quantitative PCR (RT-qPCR) Experiment

In order to determine the RNA content of the virus, a real-time fluorescent quantitative PCR was established. The virus’s total RNA was extracted using a viral RNA extraction kit. We used the one-step fluorescence quantitative PCR kit for RT-qPCR detection. The procedure was as follows: 50 °C, 15 min for reverse transcription; 95 °C, 2 min with 45 cycles of 15 s; and 60 °C, 45 s. The forward primer sequence for the influenza B virus NS2 gene amplification was 5′-TCCTCAACTCACTCTTCGAGCG-3′, the reverse primer sequence was 5′-CGGTGCTCTTGACCAAATTGG-3′, and the probe sequence was 5′-CCAATTCGAGCAGCTGAAACTGCGGT-3′. The forward primer sequence for the influenza A virus M1 gene amplification was 5′-GACCRATCCTGTCACCTCTGAC-3′, the reverse primer sequence was 5′-AGGGCATTYTTGACAAAKCGTCTA-3′, and the probe sequence was 5′-TGCAGTCCTCGCTCACTGGGCACG-3′. The forward primer sequence for human coronavirus 229E N gene amplification was 5′-TCCTTCCCGGTCTCAGTCG-3′, the reverse primer sequence was 5′-CTGTCACTTGAAGGATTCCGAG-3′, and the probe sequence was 5′-TCGCGGTCGTGGTGAATCCAAACCTCA-3′. The forward primer sequence for human coronavirus OC43 N gene amplification was 5′-TCGTTCTGGTAATGGCATCCT-3′, the reverse primer sequence was 5′-CTGATGGTTGCTGAGAGGTAG-3′, and the probe sequence was 5′-CTAAACTGGTCGGACTGATCGGCCCA-3′.

4.8. Indirect Immunofluorescence (IFA) Assay

We used IFA to detect the inhibitory effect of nanomaterials on the spread of viral infections and glass-like 96-well plates to culture MDCK cells at a density of 2 × 104 cells/well. The next day, we discarded the cell supernatant, washed twice with PBS, and added 100 μL/well of 100 TCID50/mL virus solution, which was prepared with a serum-free medium and incubated at 35 °C for 1 h. Then, we washed the plates twice with PBS, added the AgBiS2 solutions of different concentrations (0, 50, 100, and 150 μg/mL), which were prepared using the virus infection base medium, and cultured at 35 °C for 24 h. Then, we discarded the cell supernatant, added 50 μL/well of 4% paraformaldehyde to fix the cells at room temperature for 5 min, and blocked by adding 1% BSA solution at 4 °C overnight. According to the operating instructions of the influenza B virus (B/Florida/4/2006), we added the primary antibody diluted 1:800 Hemagglutinin/HA Antibody (11053-T62 kit), and then added the secondary antibody (Goat-Anti-Rabbit lgG H&L (Alexa Fluor 488) ab150077) diluted 1:500, finally added DAPI reagent and transferred the plates to an inverted fluorescence microscope for observation.

5. Conclusions

In this work, AgBiS2 nanoparticles were synthesized by doping with the bimetal method at room temperature. By comparing the synthesized Bi2S3 and Ag2S nanoparticles, the excellent inhibitory ability of AgBiS2 nanoparticles on influenza virus B is explored. The cytotoxicity test shows that the three nanoparticles have good biological safety. Then, the inhibitory effect of nanoparticles on influenza virus B was determined via RT-qPCR, TCID50, IFA, and other assays. By comparing the inhibitory effect of synthesized Bi2S3, Ag2S and AgBiS2 nanoparticles, it is found that the maximum non-toxic concentration of AgBiS2 nanoparticles is higher than that of Bi2S3 and Ag2S. In addition, AgBiS2 shows an excellent inhibitory effect on influenza virus B, and the inhibitory process occurs at the stages of influenza virus cell internalization and intracellular replication. The optimal inhibition stage of Ag2S mainly occurs in the process of virus intracellular replication, while the main inhibition stage of Bi2S3 to influenza virus occurs in the stage of virus-cell internalization and intracellular replication. It shows that bimetallic nanoparticles have better antiviral effects than single metallic materials. Furthermore, we also verify the excellent antiviral properties of AgBiS2 nanomaterials against influenza virus A and coronaviruses. In this paper, bismuth-based nanomaterials have been applied for the first time to inhibit influenza virus infection, and AgBiS2 nanoparticles with strong inhibitory effects on influenza virus A, B, and α, β coronavirus have been screened out. Therefore, with future in-depth research, application potential of bismuth-based nanomaterials in the antiviral field could be further demonstrated.

6. Patents

The authors declare that there is potential intellectual property regarding the publication of this article.

Author Contributions

Conceptualization, Y.H., R.O. and Y.M.; methodology, J.Y., J.S., Y.H., B.S., Z.Y. and L.Y.; formal analysis, J.Y., Y.H. and L.Y.; data curation, B.S., L.Y. and Y.H.; writing—original draft preparation, J.Y. and Y.H.; Writing—Reviewing Funding acquisition, Project administration, Y.H., R.O. and Y.M.; visualization, Y.H.; supervision, Y.H., R.O. and Y.M.; funding acquisition, Y.H. and R.O. All authors have read and agreed to the published version of the manuscript.

Funding

The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was supported by the Open Foundation of National Virus Resource Center (NVRC-PY-01), China National Mega-projects for Infectious Diseases (2017ZX10103009-002), Natural Science Foundation of Shanghai (19ZR1434800, 19ZR1461900), Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, Clinical research project of Shanghai Municipal Health Commission (No.201940078), and Scientific research program of Shanghai Science and Technology Commission (No.21140903200).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We thank Guiqin Wang from the Nanjing Advanced Academy of Life and Health for the helpful discussion. Many thanks to Muhammad Bakhsh and Ahsan Ali Bhutto for their diligently proofreading the article.

Conflicts of Interest

The authors declare no competing financial interests.

References

  1. Toledo-Rueda, W.; Rosas-Murrieta, N.H.; Munoz-Medina, J.E.; Gonzalez-Bonilla, C.R.; Reyes-Leyva, J.; Santos-Lopez, G. Antiviral resistance markers in influenza virus sequences in Mexico, 2000–2017. Infect. Drug Resist. 2018, 11, 1751–1756. [Google Scholar] [CrossRef] [Green Version]
  2. Ng, L.Y.; Mohammad, A.W.; Leo, C.P.; Hilal, N. Polymeric membranes incorporated with metal/metal oxide nanoparticles: A comprehensive review. Desalination 2013, 308, 15–33. [Google Scholar] [CrossRef]
  3. Sang, C.; Ma, L.; Luo, D.; Liu, H.; Li, D.; Chen, T. Designing bioresponsive metal azolate framework-based nanosystem for efficient cancer therapy. Chem. Eng. J. 2019, 371, 301–305. [Google Scholar] [CrossRef]
  4. Delaviz, N.; Gill, P.; Ajami, A.; Aarabi, M. Aptamer-conjugated magnetic nanoparticles for the efficient removal of HCV particles from human plasma samples. RSC Adv. 2015, 5, 79433–79439. [Google Scholar] [CrossRef]
  5. Allahverdiyev, A.M.; Abamor, E.S.; Bagirova, M.; Rafailovich, M. Antimicrobial effects of TiO2 and Ag2O nanoparticles against drug-resistant bacteria and leishmania parasites. Future Microbiol. 2011, 6, 933–940. [Google Scholar] [CrossRef] [PubMed]
  6. Bhatia, S.; Lauster, D.; Bardua, M.; Ludwig, K.; Angioletti-Uberti, S.; Popp, N.; Hoffmann, U.; Paulus, F.; Budt, M.; Stadtmuller, M.; et al. Linear polysialoside outperforms dendritic analogs for inhibition of influenza virus infection in vitro and in vivo. Biomaterials 2017, 138, 22–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Wang, J.; Liu, Y.; Xu, K.; Qi, Y.; Zhong, J.; Zhang, K.; Li, J.; Wang, E.; Wu, Z.; Kang, Z. Broad-spectrum antiviral property of polyoxometalate localized on a cell surface. ACS Appl. Mater. Interfaces 2014, 6, 9785–9789. [Google Scholar] [CrossRef]
  8. Sun, H.; Li, H.; Harvey, I.; Sadler, P.J. Interactions of bismuth complexes with metallothionein(II). J. Biol. Chem. 1999, 274, 29094–29101. [Google Scholar] [CrossRef] [Green Version]
  9. Andrews, P.C.; Ferrero, R.L.; Forsyth, C.M.; Junk, P.C.; Maclellan, J.G.; Peiris, R.M. Bismuth(III) Saccharinate and Thiosaccharinate Complexes and the Effect of Ligand Substitution on Their Activity against Helicobacter pylori. Organometallics 2011, 30, 6283–6291. [Google Scholar] [CrossRef]
  10. Li, H.; Wang, R.; Sun, H. Systems Approaches for Unveiling the Mechanism of Action of Bismuth Drugs: New Medicinal Applications beyond Helicobacter pylori Infection. Acc. Chem. Res. 2019, 52, 216–227. [Google Scholar] [CrossRef]
  11. Yang, N.; Tanner, J.A.; Zheng, B.J.; Watt, R.M.; He, M.L.; Lu, L.Y.; Jiang, J.Q.; Shum, K.T.; Lin, Y.P.; Wong, K.L.; et al. Bismuth complexes inhibit the SARS coronavirus. Angew. Chem. Int. Ed. Engl. 2007, 46, 6464–6468. [Google Scholar] [CrossRef]
  12. Shu, T.; Huang, M.; Wu, D.; Ren, Y.; Zhang, X.; Han, Y.; Mu, J.; Wang, R.; Qiu, Y.; Zhang, D.Y.; et al. SARS-Coronavirus-2 Nsp13 Possesses NTPase and RNA Helicase Activities That Can Be Inhibited by Bismuth Salts. Virol. Sin. 2020, 35, 321–329. [Google Scholar] [CrossRef]
  13. Yuan, S.; Wang, R.; Chan, J.F.-W.; Zhang, A.J.; Cheng, T.; Chik, K.K.-H.; Ye, Z.-W.; Wang, S.; Lee, A.C.-Y.; Jin, L.; et al. Metallodrug ranitidine bismuth citrate suppresses SARS-CoV-2 replication and relieves virus-associated pneumonia in Syrian hamsters. Nat. Microbiol. 2020, 5, 1439–1448. [Google Scholar] [CrossRef]
  14. Park, S.; Park, H.H.; Kim, S.Y.; Kim, S.J.; Woo, K.; Ko, G. Antiviral Properties of Silver Nanoparticles on a Magnetic Hybrid Colloid. Appl. Environ. Microbiol. 2014, 80, 2343–2350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ilegbusi, O.J.; Trakhtenberg, L. Synthesis and Conductometric Property of Sol-Gel-Derived ZnO/PVP Nano Hybrid Films. J. Mater. Eng. Perform. 2012, 22, 911–915. [Google Scholar] [CrossRef]
  16. Kumar, M.; Devi, P.; Shivling, V.D. Thermal stability and electrochemical properties of PVP-protected Ru nanoparticles synthesized at room temperature. Mater. Res. Express 2017, 4, 085006. [Google Scholar] [CrossRef]
  17. Kumar, S.R.; Paulpandi, M.; ManivelRaja, M.; Mangalaraj, D.; Viswanathan, C.; Kannan, S.; Ponpandian, N. An in vitro analysis of H1N1 viral inhibition using polymer coated superparamagnetic Fe3O4 nanoparticles. RSC Adv. 2014, 4, 13409–13418. [Google Scholar] [CrossRef]
  18. Ghaffari, H.; Tavakoli, A.; Moradi, A.; Tabarraei, A.; Bokharaei-Salim, F.; Zahmatkeshan, M.; Farahmand, M.; Javanmard, D.; Kiani, S.J.; Esghaei, M.; et al. Inhibition of H1N1 influenza virus infection by zinc oxide nanoparticles: Another emerging application of nanomedicine. J. Biomed. Sci. 2019, 26, 70. [Google Scholar] [CrossRef] [PubMed]
  19. Du, T.; Liang, J.; Dong, N.; Lu, J.; Fu, Y.; Fang, L.; Xiao, S.; Han, H. Glutathione-Capped Ag2S Nanoclusters Inhibit Coronavirus Proliferation through Blockage of Viral RNA Synthesis and Budding. ACS Appl. Mater. Interfaces 2018, 10, 4369–4378. [Google Scholar] [CrossRef]
  20. Jeremiah, S.S.; Miyakawa, K.; Morita, T.; Yamaoka, Y.; Ryo, A. Potent antiviral effect of silver nanoparticles on SARS-CoV-2. Biochem. Biophys. Res. Commun. 2020, 533, 195–200. [Google Scholar] [CrossRef]
  21. Tavakoli, A.; Hashemzadeh, M.S. Inhibition of herpes simplex virus type 1 by copper oxide nanoparticles. J. Virol. Methods 2020, 275, 113688. [Google Scholar] [CrossRef]
  22. Kolahalam, L.A.; Kasi Viswanath, I.V.; Diwakar, B.S.; Govindh, B.; Reddy, V.; Murthy, Y.L.N. Review on nanomaterials: Synthesis and applications. Mater. Today Proc. 2019, 18, 2182–2190. [Google Scholar] [CrossRef]
  23. Joshi, V.A.; Joshi, G.M.; Shukla, S.K. Synthesis, properties and applications of nanomaterials: A mini review. In Functionalized Nanomaterials Based Devices for Environmental Applications; Hussain, C.M., Shukla, S.K., Joshi, G.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2021; pp. 61–75. [Google Scholar]
  24. Bordiwala, R.V. Green synthesis and Applications of Metal Nanoparticles—A Review Article. Results Chem. 2023, 5, 100832. [Google Scholar] [CrossRef]
  25. Baram-Pinto, D.; Shukla, S.; Perkas, N.; Gedanken, A.; Sarid, R. Inhibition of Herpes Simplex Virus Type 1 Infection by Silver Nanoparticles Capped with Mercaptoethane Sulfonate. Bioconjugate Chem. 2009, 20, 1497–1502. [Google Scholar] [CrossRef]
  26. Li, Z.; Hu, Y.; Miao, Z.; Xu, H.; Li, C.; Zhao, Y.; Li, Z.; Chang, M.; Ma, Z.; Sun, Y.; et al. Dual-Stimuli Responsive Bismuth Nanoraspberries for Multimodal Imaging and Combined Cancer Therapy. Nano Lett. 2018, 18, 6778–6788. [Google Scholar] [CrossRef]
  27. Cui, H.; Jiang, J.; Gu, W.; Sun, C.; Wu, D.; Yang, T.; Yang, G. Photocatalytic inactivation efficiency of anatase Nano-TiO2 Sol on the H9N2 avian influenza virus. Photochem. Photobiol. 2010, 86, 1135–1139. [Google Scholar] [CrossRef] [PubMed]
  28. Imai, K.; Ogawa, H.; Bui, V.N.; Inoue, H.; Fukuda, J.; Ohba, M.; Yamamoto, Y.; Nakamura, K. Inactivation of high and low pathogenic avian influenza virus H5 subtypes by copper ions incorporated in zeolite-textile materials. Antivir. Res. 2012, 93, 225–233. [Google Scholar] [CrossRef] [PubMed]
  29. Kumar, R.; Nayak, M.; Sahoo, G.C.; Pandey, K.; Sarkar, M.C.; Ansari, Y.; Das, V.N.R.; Topno, R.K.; Bhawna; Madhukar, M.; et al. Iron oxide nanoparticles based antiviral activity of H1N1 influenza A virus. J. Infect. Chemother. 2019, 25, 325–329. [Google Scholar] [CrossRef] [PubMed]
  30. Abo-Zeid, Y.; Ismail, N.S.M.; McLean, G.R.; Hamdy, N.M. A molecular docking study repurposes FDA approved iron oxide nanoparticles to treat and control COVID-19 infection. Eur. J. Pharm. Sci. 2020, 153, 105465. [Google Scholar] [CrossRef] [PubMed]
  31. Vonnemann, J.; Sieben, C.; Wolff, C.; Ludwig, K.; Böttcher, C.; Herrmann, A.; Haag, R. Virus inhibition induced by polyvalent nanoparticles of different sizes. Nanoscale 2014, 6, 2353. [Google Scholar] [CrossRef] [PubMed]
  32. Yang, J.; Yue, L.; Yang, Z.; Miao, Y.; Ouyang, R.; Hu, Y. Metal-Based Nanomaterials: Work as Drugs and Carriers against Viral Infections. Nanomaterials 2021, 11, 2129. [Google Scholar] [CrossRef] [PubMed]
  33. Du, J.; Gu, Z.; Yan, L.; Yong, Y.; Yi, X.; Zhang, X.; Liu, J.; Wu, R.; Ge, C.; Chen, C.; et al. Poly(Vinylpyrollidone)- and Selenocysteine-Modified Bi2Se3 Nanoparticles Enhance Radiotherapy Efficacy in Tumors and Promote Radioprotection in Normal Tissues. Adv. Mater. 2017, 29, 1701268. [Google Scholar] [CrossRef] [PubMed]
  34. Tavakoli, A.; Ataei-Pirkooh, A.; Mm Sadeghi, G.; Bokharaei-Salim, F.; Sahrapour, P.; Kiani, S.J.; Monavari, S.H. Polyethylene glycol-coated zinc oxide nanoparticle an efficient nanoweapon to fight against herpes simplex virus type 1. Nanomedicine 2018, 13, 2675–2690. [Google Scholar] [CrossRef] [PubMed]
  35. Baram-Pinto, D.; Shukla, S.; Gedanken, A.; Sarid, R. Inhibition of HSV-1 attachment, entry, and cell-to-cell spread by functionalized multivalent gold nanoparticles. Small 2010, 6, 1044–1050. [Google Scholar] [CrossRef]
  36. Reed, L.J.; Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 1938, 27, 493–497. [Google Scholar] [CrossRef]
Figure 1. TEM images of nanoparticles (a) AgBiS2, (b) Bi2S3, and (c) Ag2S; XRD diffraction patterns of nanoparticles (d) AgBiS2, (e) Bi2S3, and (f) Ag2S.
Figure 1. TEM images of nanoparticles (a) AgBiS2, (b) Bi2S3, and (c) Ag2S; XRD diffraction patterns of nanoparticles (d) AgBiS2, (e) Bi2S3, and (f) Ag2S.
Ijms 24 10223 g001
Figure 2. The mapping spectrum of Bi, S, and Ag of AgBiS2 nanoparticles.
Figure 2. The mapping spectrum of Bi, S, and Ag of AgBiS2 nanoparticles.
Ijms 24 10223 g002
Figure 3. (a) Size distributions, (b) Zeta potential, (c) Infrared spectroscopy analysis of AgBiS2, Bi2S3, and Ag2S; (d) thermogravimetric analysis of AgBiS2, and Bi2S3.
Figure 3. (a) Size distributions, (b) Zeta potential, (c) Infrared spectroscopy analysis of AgBiS2, Bi2S3, and Ag2S; (d) thermogravimetric analysis of AgBiS2, and Bi2S3.
Ijms 24 10223 g003
Figure 4. Cytotoxicity tests of (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles.
Figure 4. Cytotoxicity tests of (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles.
Ijms 24 10223 g004
Figure 5. RT-qPCR analysis of (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles’ direct inactivating effects on influenza virus B. The inhibitory effect of the control group was 0% which was not shown in the figure.
Figure 5. RT-qPCR analysis of (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles’ direct inactivating effects on influenza virus B. The inhibitory effect of the control group was 0% which was not shown in the figure.
Ijms 24 10223 g005
Figure 6. RT-qPCR analysis of (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles and influenza virus B co-cultivation of the inhibitory effects. The inhibitory effect of the control group was 0% which was not shown in the figure.
Figure 6. RT-qPCR analysis of (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles and influenza virus B co-cultivation of the inhibitory effects. The inhibitory effect of the control group was 0% which was not shown in the figure.
Ijms 24 10223 g006
Figure 7. RT-qPCR analysis of the effects on influenza B of the nanoparticle preculture inhibition group. (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles. The inhibitory effect of the control group was 0% which was not shown in the figure.
Figure 7. RT-qPCR analysis of the effects on influenza B of the nanoparticle preculture inhibition group. (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles. The inhibitory effect of the control group was 0% which was not shown in the figure.
Ijms 24 10223 g007
Figure 8. RT-qPCR analysis of the inhibitory effects on influenza B of the virus post-infection inhibition group. (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles. The inhibitory effect of the control group was 0% which was not shown in the figure.
Figure 8. RT-qPCR analysis of the inhibitory effects on influenza B of the virus post-infection inhibition group. (a) AgBiS2, (b) Ag2S, and (c) Bi2S3 nanoparticles. The inhibitory effect of the control group was 0% which was not shown in the figure.
Ijms 24 10223 g008
Figure 9. The TCID50 method is used to detect the influenza B virus titer of AgBiS2 (a) direct inactivating group, (b) co-cultivation inhibition group, (c) nanoparticle preculture inhibition group, and (d) virus post-infection inhibition group.
Figure 9. The TCID50 method is used to detect the influenza B virus titer of AgBiS2 (a) direct inactivating group, (b) co-cultivation inhibition group, (c) nanoparticle preculture inhibition group, and (d) virus post-infection inhibition group.
Ijms 24 10223 g009
Figure 10. The TCID50 method is used to detect the influenza B virus titer of Ag2S (a) direct inactivating group, (b) co-cultivation inhibition group, (c) nanoparticle preculture inhibition group, and (d) virus post-infection inhibition group.
Figure 10. The TCID50 method is used to detect the influenza B virus titer of Ag2S (a) direct inactivating group, (b) co-cultivation inhibition group, (c) nanoparticle preculture inhibition group, and (d) virus post-infection inhibition group.
Ijms 24 10223 g010
Figure 11. The TCID50 method is used to detect the influenza B virus titer of Bi2S3. (a) the direct inactivating group, (b) co-cultivation inhibition group, (c) nanoparticle preculture inhibition group, and (d) virus post-infection inhibition group.
Figure 11. The TCID50 method is used to detect the influenza B virus titer of Bi2S3. (a) the direct inactivating group, (b) co-cultivation inhibition group, (c) nanoparticle preculture inhibition group, and (d) virus post-infection inhibition group.
Ijms 24 10223 g011
Figure 12. The indirect immunofluorescence assay detects the influenza B virus HA expression affected by AgBiS2 at the concentration of 0, 50, and 150 μg/mL, and the virus post-infection group are treated for 24 h. DAPI is a nuclear dye, and T-62 is a fluorescent primary antibody to the influenza virus HA antigen.
Figure 12. The indirect immunofluorescence assay detects the influenza B virus HA expression affected by AgBiS2 at the concentration of 0, 50, and 150 μg/mL, and the virus post-infection group are treated for 24 h. DAPI is a nuclear dye, and T-62 is a fluorescent primary antibody to the influenza virus HA antigen.
Ijms 24 10223 g012
Figure 13. RT-qPCR analysis of the inhibitory effects of the virus post-infection inhibition group on influenza virus A (a), β coronavirus OC43 (b) and α coronavirus 229E, and (c) by AgBiS2 nanoparticles.
Figure 13. RT-qPCR analysis of the inhibitory effects of the virus post-infection inhibition group on influenza virus A (a), β coronavirus OC43 (b) and α coronavirus 229E, and (c) by AgBiS2 nanoparticles.
Ijms 24 10223 g013
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, J.; Yue, L.; Shen, B.; Yang, Z.; Shao, J.; Miao, Y.; Ouyang, R.; Hu, Y. Exploring the Inhibitory Effect of AgBiS2 Nanoparticles on Influenza Viruses. Int. J. Mol. Sci. 2023, 24, 10223. https://doi.org/10.3390/ijms241210223

AMA Style

Yang J, Yue L, Shen B, Yang Z, Shao J, Miao Y, Ouyang R, Hu Y. Exploring the Inhibitory Effect of AgBiS2 Nanoparticles on Influenza Viruses. International Journal of Molecular Sciences. 2023; 24(12):10223. https://doi.org/10.3390/ijms241210223

Chicago/Turabian Style

Yang, Junlei, Lihuan Yue, Bei Shen, Zhu Yang, Jiang Shao, Yuqing Miao, Ruizhuo Ouyang, and Yihong Hu. 2023. "Exploring the Inhibitory Effect of AgBiS2 Nanoparticles on Influenza Viruses" International Journal of Molecular Sciences 24, no. 12: 10223. https://doi.org/10.3390/ijms241210223

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop