Next Article in Journal
Parasitic Granulomatous Dermatitis Caused by Pelodera spp. in Buffalo on Marajó Island, Pará
Previous Article in Journal
Use of Cracker Residue in the Diet of Dairy Heifers: Impacts on Animal Health, Ruminal Fatty Acids Profile, Digestibility, Weight Gain, and Economic Viability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Analysis of the Immunogenicity of African Swine Fever F317L Protein and Screening of T Cell Epitopes

1
Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
2
Animal Science and Technology College, Beijing University of Agriculture, Beijing 100193, China
*
Author to whom correspondence should be addressed.
Animals 2024, 14(9), 1331; https://doi.org/10.3390/ani14091331
Submission received: 9 March 2024 / Revised: 15 April 2024 / Accepted: 23 April 2024 / Published: 29 April 2024
(This article belongs to the Section Veterinary Clinical Studies)

Abstract

:

Simple Summary

The African swine fever virus (ASFV) is a linear double-stranded DNA virus characterized by a complex immune escape mechanism that has caused substantial economic losses to the global swine industry. An in-depth study of ASFV has revealed that screening for protective antigens and activating cellular immunity is essential for the development of ASF vaccines. In this study, we sought to examine the immunogenicity of the F317L protein of ASFV in mice while also identifying potential active T-cell epitopes in order to provide a basis for the functional study of the F317L protein, in addition to a reference for the subsequent development of ASF vaccines.

Abstract

The African swine fever virus (ASFV) encodes numerous proteins characterized by complex immune escape mechanisms. At present, the structure and function of these proteins, including the F317L protein, have yet to be fully elucidated. In this study, we examined the immunogenicity of the F317L protein. Mice were subcutaneously immunized with the F317L protein using initial and subsequent booster doses, and, at the 28th day post-treatment, we assessed the humoral and cellular immune responses of mice. The F317L protein stimulated production of specific antibodies and activated humoral immune responses. In addition, F317L stimulated the production of large amounts of IFN-γ by splenic lymphocytes, thereby activating cellular immune responses. Using informatics technology, we predicted and synthesized 29 F317L protein T cell epitopes, which were screened using IFN-γ ELISpot. Among these, the F25 (246SRRSLVNPWT255) peptide was identified as having a stronger stimulatory effect than the full-length protein. Collectively, our findings revealed that the ASFV F317L protein can stimulate both strong humoral and cellular immunity in mice, and that the F25 (246SRRSLVNPWT255) peptide may be a potential active T cell epitope. These findings will provide a reference for further in-depth studies of the F317L protein and screening of antigenic epitopes.

1. Introduction

African swine fever (ASF) is an acute highly contactable infectious disease caused by the African swine fever virus (ASFV) that infects domestic and wild pigs [1]. Highly virulent strains of the virus can cause hyperacute and acute disease, resulting in 100% mortality in pigs within a short period of time after infection [1,2]. ASF is classified as a legally reportable animal disease by the World Health Organization, and it is also classified as a Class I animal disease in China. Since the first reported outbreak of ASF in Shen-yang City, Liaoning Province, China, in August 2018, ASF has spread rapidly to most parts of China, causing considerable economic losses to the domestic pig farming industry.
The relatively large ASFV genome encodes between 150 and 200 proteins [3]. The virion consists of five concentric layers, extending outward from the central inner core (via the inner core shell, inner membrane, and capsid) to the exterior vesicle, each of which comprises distinct protein types [4,5]. These mainly include structural and immunoregulatory proteins [6], including the inner membrane proteins p54, p17, and p30; coat proteins p72 and p49; and vesicle proteins CD2v and p12. Among these, it has previously been confirmed that the ASFV p54, p72, and p30 proteins can induce the production of neutralizing antibodies in experimental animals [7,8]. Attack protection experiments revealed that immunizing pigs with these three proteins failed to protect these pigs, indicating that the induced production of neutralizing antibodies was insufficient in regard to conferring immunity [9].
The findings of further in-depth studies on the virus have revealed that the speed and efficacy of the neutralizing antibody response show considerable variation [7]; additionally, there may be differences in the neutralization mechanism of the antibodies elicited by different proteins. Consequently, the associations between the neutralizing antibodies and immunity protection against ASFV are in need of further experimental verification [10,11]. In this regard, it has been established that ASFV specific antibodies alone are insufficient for protecting against ASFV infection, while CD8+ lymphocyte subsets have been demonstrated to play an important role in protective immunity against ASFV [12,13]. Furthermore, a close association between cellular immune responses and protective responses has been identified in inbred pigs [14], and Argilaguet et al. [15] have described a DNA vaccination strategy in which 33% of the immunized pigs were immunoprotected, with this protective effect being attributed to an induction of CD8+ T cells. The findings of this study thus indicate that T cell immunity plays an important role in immunity protection. However, while, at present, ASFV protective antigens have yet to be identified, given that peptides induce protective responses, further screening and characterization of antigenic T-cell epitopes [16] are considered to be essential for the development of effective ASF vaccines.
On the basis of a literature search, we found that the late F317L protein of ASFV has been demonstrated to have immunosuppressive effects by impairing activation of the NF-kB pathway, thereby disrupting NF-kB activity [17]. In a further study, the authors analyzed the reactogenicity of the F317L protein and predicted that it has six B-cell antigenic epitopes [18]. The findings of such studies have thus provided evidence to indicate that ASFV F317L is a potential immunogenic antigen. Consequently, in this study, we sought to analyze F317L, and, accordingly, found that this protein can elicit both humoral and cellular immunity in experimental mice. In addition, we performed in silico prediction and subsequent synthesis of F317L T-cell epitopes, successfully identifiing an epitope using the IFN-γ ELISpot method. Our findings in this study will provide a basis for further in-depth studies of ASFV F317L immunogenicity, as well as a technical reference for the subsequent functional studies of the F317L protein and identification of T-cell active epitopes.

2. Materials and Methods

2.1. Protein and Cells

ASFV F317L and p72 recombinant proteins were prepared and preserved in this experiment. Experimentally obtained mice splenic lymphocytes were cultured with RPMI 1640 medium (Gibco, Grand Island, NE, USA, Cat. No#. 11875093) containing 10% Fetal Bovine Serum (FBS) (Gibco, Grand Island, NE, USA, Cat. No#. A31604-01) and 100 units/mL penicillin-streptomycin (Gibco, Grand Island, NE, USA, Cat. No#. 15140-122).

2.2. Animals

A total of 18 SPF (Specific pathogen Free) grade 6–8-weeks-old female C57BL/6J mice were purchased from Beijing Viton Lihua Laboratory Animal Technology Co., Ltd. (Beijing, China) and kept in a negative-pressure barrier environment in the Laboratory Animal Center of the Beijing Institute of Animal Husbandry and Veterinary Research, Chinese Academy of Agricultural Sciences. All experimental activities were approved by the Experimental Animal Welfare Ethics Committee of Beijing Institute of Animal Science and Veterinary Medicine, Chinese Academy of Agricultural Sciences (No. IAS2022-157).

2.3. Animal Experiments

To assess the immunogenicity of the ASFV F317L protein in mice, female SPF C57BL/6J mice were immunized with recombinant F317L protein that was previously expressed and identified. In detail, six mice in each group were subcutaneously immunized at a dose of 10 μg with prime and booster immunizations at Day 0 and Day 21, respectively. As the immunized group, positive and negative controls, mice were administered with F317L protein, p72 protein, and phosphate-buffered saline (PBS), respectively. Subsequently, serum was collected from the orbits of mice on Days 7, 14, 21, and 28, and the spleens of mice were extracted 7 days after the booster immunization (Figure 1). The splenic lymphocytes obtained from three mice in the F317L protein group were used for IFN-γ ELISpot Assays and flow cytometry assay, while another three mice were used for peptide screening.

2.4. Mice Splenic Lymphocytes Isolation

Mice splenic lymphocytes were isolated at Day 28 post-immunization. After being euthanized, the spleens from mice of each group (F317L protein immunized group, p72 protein positive and PBS negative controls) were taken out to make splenocyte suspension, and mice splenic lymphocytes were isolated by a Mouse Splenic Lymphocyte Isolation Kit (Solarbio, Beijing, China, Cat. No#. P8860). The middle milky white lymphocyte layer was washed twice and centrifuged 5 min under 1000 rpm; then, the splenic lymphocytes were suspended in RPMI1640 complete culture medium.

2.5. IFN-γ ELISpot Assays

In order to detect the cellular immunity level of spleen cells in immunized mice, IFN-γ ELISpot assays were performed using a Mouse IFN-γ precoated ELISpot kit (Dakewe, Shenzhen, China, Cat. No#2210006) according to the manufacturer’s instructions. Isolated splenic lymphocytes from mice immunized with F317L protein were seeded into pre-coated 96-well IFN-γ ELISpot plates at a density of 2 × 106/mL per well, three replicate per mice, then stimulated with 100 ng of F317L protein. At the same time, the splenic lymphocytes from p72 protein immunized mice used as positive control and stimulated with 100 ng of p72 protein, and those obtained from mice received no stimulation used as negative control. All positive controls in each group were used with 5 ng PMA and 100 ng Ionomycin of positive stimulant (Dakewe, Shenzhen, China, Cat. No#2030421). Then, the plates were incubated in a culture incubator at 37 °C, 5% CO2 for 24 h, after which the cells were lysed for 10 min with pre-cooled deionized water at 4 °C and washed with washing buffer six times, Biotinylated Antibody was added, and plates were subsequently incubated at 37 °C for 1 h, then washed with washing buffer six times. Streptavidin-HRP was added and incubated at 37 °C for 1 h and washed with washing buffer six times. AEC color development solutions were added, kept in the dark or 30 min at room temperature (RT), and then washed with washing buffer six times. Finally, the plates were dried naturally at RT, and the spot-forming Cells (SFCs) were counted using an IRIS Mabtech ELISpot/FluoroSpot plate reader (Mabtech, Stockholm, Sweden).

2.6. CD4+/CD8+ T Cell Subclass Ratio

Splenic lymphocyte CD4+/CD8+ subclass analysis was performed using flow cytometry. An amount of 97 µL of cell staining buffer (BioLegend, San Diego, CA, USA, Cat. No#420201) and 0.5 µL of TruStain FcXTM PLUS (anti-mouse CD16/32) (BioLegend, Cat. No#156604) antibody were transferred to 1.5 mL Eppendorf (EP) tubes containing 1.5 × 106 cells, mixed softly, and then used to stain the cells for 10 min at RT in the dark. Thereafter, 0.5 µL of FITC anti-mouse CD3ε antibody (BioLegend, San Diego, CA, USA, Cat. No#100204), 1.25 µL of PerCP/Cyanine5.5 anti-mouse CD8α antibody (BioLegend, San Diego, CA, USA, Cat. No#100734), and 1.25 µL of PE anti-mouse CD4 antibody (BioLegend, San Diego, CA, USA, Cat. No#100512) were added to the EP tubes, mixed softly, and then used to stain the cells for 30 min at 4 °C in the dark. Then, the EP tubes were centrifuged at 200× g for 5 min, and the supernatant was discarded. Next, 1 mL of PBS was added to make a cell suspension, centrifuged at 4 °C, 200× g for 5 min, and the supernatant was then removed. Cells were centrifuged twice, and 250 µL of Cell Staining Buffer was added for cell suspension. Finally, the cells were analyzed by FACSVerse flow cytometer (BD Bioscience, Franklin Lakes, NJ, USA), and all dates were analyzed using FlowJoV_10 software.

2.7. Indirect ELISA

The F317L and p72 protein were diluted to 2 µg/mL with carbonate buffer solution (pH 9.6). An amount of 100 µL aliquots were added to each well in 96-well ELISA plates and incubated for 14 h overnight at 4 °C. The plates were sealed with 300 µL of 5% skimmed milk at 37 °C for 2 h, then washed with PBS with 0.01% Tween-20 (PBST), and the serum (1:3200) was then added and incubated at 37 °C for 1 h. After washing with PBST, HRP-conjugated goat anti-mouse IgG (1:5000) (Abcam, Cambridge, UK, Cat. No#ab6789) was added and incubated at 37 °C for 1 h. The plates were incubated with 100 µL KPL SureBlueTM TMB Microwell Peroxidase Substrate (KPL, Milford, MA, USA, Cat. No#5120-0077) at RT for 15 min, and 100 µL of 1 M HCl was added to terminate the reaction, after which the results were expressed as optical density (OD) at 450 nm. Values were assessed based on a threshold indirect ELISA value optimized in the laboratory. Samples with an experimental well/blank well ratio OD450 value ≥ 2.1 specifically were deemed to be positive, whereas those with a value < 2.1 were assessed to be negative.

2.8. Cytokine Assays

The levels of IL2, IL4, IFN-γ, and TNF-α in mouse sera were determined using a Q-Plex™ Mouse HS kit (Cayman Chemical, Michigan, USA, Cat, No#39970) according to the manufacturer’s instructions. Briefly, 30 µL of serum samples were added to the wells of plates, and the plates were covered with a plate seal and incubated at RT (20–25 °C). Thereafter, the wells were washed three times, followed by the sequential addition of 50 µL of Detection Mix, Streptavidin-HRP, and substrate with incubations at RT for 2 h, 20 min, and 5 min, respectively. The plates were then placed in the Q-View Imager (Quansys Biosciences, Utah, USA), with the images thus obtained being processed using Q-View software 3.0, resulting in the generation of charts, concentrations, and statistics.

2.9. Prediction of T Cell Epitopes of ASFV F317L Protein

The T cell antigenic epitope region of the F317L protein amino acid sequence (GenBank ID: UFD97811.1, Beijing, China) was analyzed by IEDB server V2.22 (https://www.iedb.org/, accessed on 4 September 2023) and NetMHCpan-4.1 server (https://services.healthtech.dtu.dk/services/NetMHCpan-4.1, accessed on 4 September 2023). Peptide affinity was assessed based on a percentile rank, with a lower rank% value being taken to be indicative of a stronger peptide affinity. The combined results yielded 29 high-affinity peptides (Table 1), the predicted sequences of which were chemically synthesized by GenScript Ltd. (Nanjing, China). These peptides, for which the purity was greater than 95%, were dissolved in the recommended solvents (ultrapure water, formic acid, DMSO, paminoacetic acid) at a concentration of 2 mg/mL.

2.10. F317L Protein T Cell Epitope Screening

The splenic lymphocytes of immunized mice were used for T-cell epitopes screening, and were stimulated by the epitopes with 10 μg per well. The negative control was stimulated with solvents, and the positive control was stimulated with 5 ng PMA and 100 ng Ionomycin of positive stimulant. All the treated cells were incubated at 37 °C, 5% CO2 for 20 h. Then, the following steps processed as described in Section 2.6.

2.11. Statistical Analysis

Statistical analysis and plotting were performed using GraphPad Prism 8.0 software. Comparisons between groups were performed using a student’s t-test. Data were expressed as mean ± standard deviation (SD). The p-value obtained from each analysis was indicated in the graph, and p-values < 0.05 were considered statistically significant.

3. Results

3.1. The F317L Protein Stimulates Splenic Lymphocytes to Secrete IFN-γ

IFN-γ ELISpot assays were performed on Day 7 after booster immunization to detect the cellular immunity level of the spleen cells of immunized mice. The results revealed that the splenic lymphocytes of F317L protein and p72 protein in immunized mice produced higher levels IFN-γ than those in the negative control group (p < 0.001) when stimulated with 100 ng of F317L protein and p72 protein, respectively (Figure 2). The numbers of IFN-γ spots stimulated with the F317L protein were significantly higher than those in the p72 positive control group (p < 0.01). These findings accordingly indicated that the F317L protein could effectively activate the cellular immunity and stimulate the secretion of IFN-γ by the spleen lymphocytes of immunized mice.

3.2. The F317L Protein Can Induce an Increase in the CD4+/CD8+ Ratio of Mouse Splenic Lymphocytes and Promote Differentiation to CD4+ T Cell Subsets

Flow cytometry analysis of T lymphocyte subpopulations in each group of mice revealed significant differences between the F317L protein group and the negative (p < 0.01) and/or p72 positive control groups (p < 0.05) with respect to the CD4+/CD8+ ratio of splenic lymphocytes (Figure 3), which indicated that the F317L protein could stimulate the splenic lymphocytes of mice to differentiate toward CD4+ T cells and activate immune response in vivo.

3.3. The F317L Protein Induced and the Secretion of High Specific Antibodies in Humoral Immunity in Immunized Mice

Indirect ELISA assays were performed to detect specific antibody levels on Days 7, 14, 21, and 28 post-immunization. The results shows that specific antibodies were detected as soon as Day 7, and the antibody levels increased gradually with time, being maintained at an elevated level during the remaining period (Figure 4). Additionally, the specific antibody levels in the F317L protein group were lower than those in the p72 positive control group, although they still remained at a high level. These findings indicated that the F317L protein could induce the humoral immunity.

3.4. F317L Protein Promoted the Secretion of Immune-Related Cytokines (IL-2, IL-4, IFN-γ, and TNF-α) in Immunized Mice

The serum cytokine contents measured on Day 21 after primer immunization and 7 days after the booster immunization revealed that the levels of IL2, IL4, IFN-γ, and TNF-α cytokines in F317L protein immunized mice were higher than those in the negative control group mice (Figure 5). Additionally, the level of Day 28 post-inoculation was higher than Day 21 (p < 0.001). The results indicated that F317L protein promoted the secretion of immune-related cytokines (IL-2, IL-4, IFN-γ, and TNF-α) in immunized mice both in the initial and booster immunization stage, and the level of cytokines produced after secondary immunization was higher than the primer, with the difference being very significant (p < 0.001).

3.5. Peptide(246−255aa) Is Potential T Cell Active Epitopes of F317L Protein

To identify the T cell epitope of the F317L protein, we predicted and subsequently synthesized 29 peptides of 9~11 amino acids, which were subjected to IFN-γ ELISpot analyses by co-incubating cells exposed to different stimuli. The results obtained on the 7th day following booster immunization with the F317L protein revealed different numbers of IFN-γ spots in cells treated with the different peptides. In particular, the number of spots per SFC/105 cells treated with the F25 (246SRRSLVNPWT255) peptide was found to be significantly higher than that of the F317L protein (Figure 6). Accordingly, F25 (246SRRSLVNPWT255) peptide was identified as a putative T-cell epitope by IFN-γ ELISpot assays, which can induce a strong immune response.

4. Discussion

Despite the fact that African swine fever has been endemic for over a century, to date, no effective and safe vaccine against it has been successfully developed. ASFV is characterized by a large genome containing between 160 and 175 open reading frames [19] that encode numerous proteins that contribute to the initiation of complex immune escape mechanisms [5]. Although inactivated vaccines produce strong humoral immunity, they tend to be ineffective in stimulating cellular immune responses, and therefore fail to provide immune protection [20,21,22]. However, vector-based vaccines constructed using multiple types of viral antigens (including p220, p72, p15, B602L, p62, p32, p54, EP153R, p12, and CD2v [23,24,25,26]) that produce good specific antibody responses, and a certain degree of cellular immunity, can provide partial protection [9,27,28,29,30]. Moreover, safety remains a major concern for weak vaccines [31], and, although gene deletion-based vaccines can provide partial protection [32], there is a need to identify suitable target genes and combinations of deletions [33]. Currently, the ASF subunit vaccine can produce better humoral immunity [34] but cannot stimulate efficient cellular immunity. Consequently, identifying effective protective antigens and establishing approaches for activating cellular immunity [10,31] are of particular importance for vaccine development [16].
The results in this study provided evidence to indicate that F317L protein may serve as a potential immunogenic antigen of ASFV. Given that the ASFV p72 protein is a major component of the viral capsid, which is an important criterion for virus typing [35] and also an important antigen of ASFV, we used this protein as a positive control in this study. The results of indirect ELISA analyses indicated that the F317L protein can activate humoral immunity, which can stimulate mice to produce large numbers of specific antibodies. In addition, we used the sensitive ELISpot assay to detect IFN-γ [36], which can serve as an important indicator for assessing the activation of cellular immunity. The results indicated that the F317L protein can significantly stimulate the production of IFN-γ and activated cellular immune responses in F317L protein immunized mice compared to the p72 protein positive control. In addition, flow cytometry analyses revealed a significant increase in the CD4+ T percentage of T lymphocytes stimulated by the F317L protein compared with that of p72 protein positive control. Cytokines are important mediators of the immune response, with Th1 cells mainly secreting IL-2, IFN-γ, and TNF-α, and Th2 cells mainly secreting IL-4. Cytotoxic T lymphocytes (CTLs) can play important roles in combatting infection via the secretion of cytokines such as IFN-γ and TNF-α [37], the former of which, secreted by cells in response to viral infection, can enhance natural killer (NK) cell and macrophage activity, while porcine IFN-γ has been established to reduce ASFV replication in porcine macrophages in vitro [38]. The results of our cytokine assays in this study revealed significantly elevated levels of immune-related cytokines (IL-2, IL-4, IFN-γ, and TNF-α) in response booster immunization, which also provides evidence to indicate that these immunized mice produced both humoral and cellular immune responses.
Studies in recent years have shown that ASFV-neutralizing antibodies alone do not provide adequate protection from infection [9,21,39]. Both NK and CTL cells detect and attack pathogen-infected cells and play important roles in mounting a resistance to ASFV infection [40,41]. The findings of some studies have indicated that the CD8α+ T lymphocyte response is closely associated with immunity protection [10,15,33,42], providing evidence that cellular immunity is a key facet of the host response to ASFV infection. Cellular immunity is mediated primarily via an endogenous delivery pathway, which implies that active T cell epitopes with high affinity for SLA (swine leukocyte antigen) molecules play an essential role in activating cellular immune responses. In this regard, Sun et al. [43] coupled multiple T cell epitopes of ASFV and constructed a nano-vaccine, which was found to be highly immunogenic and produced strong humoral and cellular immune responses in mice following immunization. Given that antigenic peptides can bind directly to SLA molecules and efficiently stimulate strong humoral and cellular immunity, the screening and characterization of antigenic peptide libraries have attracted considerable attention in recent years. Seventeen peptides derived from 15 different antigens screened using SLAI were found to stimulate strong humoral and cellular immune responses in pigs immunized for 21 days [44]. Lymphocyte peptide libraries for anti-ASFV immunization were screened using surviving porcine peripheral blood mononuclear cells (PBMCs) and randomly divided into sub-peptide Libraries A and B for immunization, with both groups being found to stimulate the production of strong cellular immune responses [10]. The T cell epitope of the CD2V single antigen, which promotes CD8+ T cell metabolism, was found to induce significantly greater viral clearance than whole-protein immunization [45].
The findings of these studies have highlighted that ASFV specific T cell epitopes are important factors in stimulating the generation of efficient cellular immunity, and, consequently, the prediction and screening of T cell epitopes are considered key steps in the development of novel subunit vaccines. The techniques currently employed for predicting and screening T cell epitopes are relatively well developed, with in silico prediction, IFN-γ ELISpot, and immunepeptidomics approaches being commonly adopted [16,36,46]. In the present study, we used relevant software (IEDB server V2.22 and NetMHCpan-4.1 server) to predict and subsequently synthesize 29 F317L protein T-cell epitopes, and we also screened for active T cell epitopes using an IFN-γ ELISpot assay. Among these, we demonstrated that peptide F25 (246SRRSLVNPWT255) could stimulate mouse splenic lymphocytes to produce large amounts of IFN-γ, and, in this regard, it proved to be more potent than the stimulatory effects of the full-length F317L protein. We firstly and systematically evaluated the humoral immunity and cellular immunity of ASFV F317L protein, further identifying the T cell epitope of this protein, and the results thus provide evidence to indicate that the F25 (246SRRSLVNPWT255) peptide is a putative T cell-dominant epitope. Given that data obtained using pigs can provide a more reliable indication of the immunogenicity of ASFV proteins, in further studies we will seek to validate the effects of this F317L protein T cell epitope on surviving porcine PBMCs, which could accordingly provide a basis for the development of a novel ASF subunit vaccine based on a peptide library of antigenic epitopes, with the aim being to provide effective protection.

5. Conclusions

In this study, we conducted animal experiments to assess the immunogenicity of the African swine fever virus F317L protein, which was found to stimulate both humoral and cellular immune responses in mice. In addition, by performing IFN-γ ELISpot analyses, we identified a potentially active T cell epitope F25 (246SRRSLVNPWT255). Collectively, our findings indicate that the F317L protein has good immunogenicity in mice. Further studies will focus on evaluating the immunogenicity of this protein and the elicited immune protection in pigs, the findings of which will provide a reference for the screening of target antigens of African swine fever vaccines.

Author Contributions

Conceptualization, Y.H. (Ying Huang) and H.J.; methodology, Y.H. (Ying Huang), H.J. and W.Z.; software, C.T.; formal analysis, Y.H. (Ying Huang) and W.Z.; investigation, Y.H. (Ying Huang), W.Z., Y.C., Z.W. and Y.H. (Yuheng He); resources, Y.H. (Yuheng He); data curation, Y.H. (Ying Huang) and W.Z.; Writing—Original draft preparation, Y.H. (Ying Huang); Writing—Review and editing, H.J.; visualization, Y.H. (Ying Huang) and Z.W.; supervision, W.Z. and Z.P.; project administration, H.J. and H.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key Research and Development Program of China, grant number 2021YFD1801200, and the Yunnan Key Research and Development Project, grant number 202103AC100001, the Earmarked Fund for CARS, grant number CARS36, and the Agricultural Science and Technology Innovation Program, grant number ASTIP-IAS-11.

Institutional Review Board Statement

The animal study protocol was approved by the Ethics Committee of Experimental Animal Welfare of CAAS (protocol code IAS2022-157 and date of approval No. 2021YFD1801200).

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Acknowledgments

First: I would like to thank the organizations that gave financial support to this study. Secondly, I would like to thank my supervisor, researcher Jia Hong, for the patient guidance given to me. In addition, I would like to thank my fellow students for their companionship and help in the preparation of this thesis. Finally, I would like to thank all my friends who have read this article, and I hope that you can provide valuable suggestions for this article, which are sincerely appreciated.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of the data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Dixon, L.K.; Sun, H.; Roberts, H. African swine fever. Antiviral Res. 2019, 165, 34–41. [Google Scholar] [CrossRef] [PubMed]
  2. Pietschmann, J.; Guinat, C.; Beer, M.; Pronin, V.; Tauscher, K.; Petrov, A.; Keil, G.; Blome, S. Course and transmission characteristics of oral low-dose infection of domestic pigs and European wild boar with a Caucasian African swine fever virus isolate. Arch. Virol. 2015, 160, 1657–1667. [Google Scholar] [CrossRef] [PubMed]
  3. Li, Z.; Chen, W.; Qiu, Z.; Li, Y.; Fan, J.; Wu, K.; Li, X.; Zhao, M.; Ding, H.; Fan, S.; et al. African Swine Fever Virus: A Review. Life 2022, 12, 1255. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, G.; Xie, M.; Wu, W.; Chen, Z. Structures and Functional Diversities of ASFV Proteins. Viruses 2021, 13, 2124. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, Y.; Kang, W.; Yang, W.; Zhang, J.; Li, D.; Zheng, H. Structure of African Swine Fever Virus and Associated Molecular Mechanisms Underlying Infection and Immunosuppression: A Review. Front. Immunol. 2021, 12, 715582. [Google Scholar] [CrossRef] [PubMed]
  6. Alejo, A.; Matamoros, T.; Guerra, M.; Andres, G. A Proteomic Atlas of the African Swine Fever Virus Particle. J. Virol. 2018, 92, e01293-18. [Google Scholar] [CrossRef] [PubMed]
  7. Escribano, J.M.; Galindo, I.; Alonso, C. Antibody-mediated neutralization of African swine fever virus: Myths and facts. Virus Res. 2013, 173, 101–109. [Google Scholar] [CrossRef] [PubMed]
  8. Gomez-Puertas, P.; Rodriguez, F.; Oviedo, J.M.; Ramiro-Ibanez, F.; Ruiz-Gonzalvo, F.; Alonso, C.; Escribano, J.M. Neutralizing antibodies to different proteins of African swine fever virus inhibit both virus attachment and internalization. J. Virol. 1996, 70, 5689–5694. [Google Scholar] [CrossRef]
  9. Neilan, J.G.; Zsak, L.; Lu, Z.; Burrage, T.G.; Kutish, G.F.; Rock, D.L. Neutralizing antibodies to African swine fever virus proteins p30, p54, and p72 are not sufficient for antibody-mediated protection. Virology 2004, 319, 337–342. [Google Scholar] [CrossRef]
  10. Netherton, C.L.; Goatley, L.C.; Reis, A.L.; Portugal, R.; Nash, R.H.; Morgan, S.B.; Gault, L.; Nieto, R.; Norlin, V.; Gallardo, C.; et al. Identification and Immunogenicity of African Swine Fever Virus Antigens. Front. Immunol. 2019, 10, 1318. [Google Scholar] [CrossRef]
  11. Rock, D.L. Thoughts on African Swine Fever Vaccines. Viruses 2021, 13, 943. [Google Scholar] [CrossRef] [PubMed]
  12. Takamatsu, H.H.; Denyer, M.S.; Lacasta, A.; Stirling, C.M.; Argilaguet, J.M.; Netherton, C.L.; Oura, C.A.; Martins, C.; Rodriguez, F. Cellular immunity in ASFV responses. Virus Res. 2013, 173, 110–121. [Google Scholar] [CrossRef] [PubMed]
  13. Attreed, S.E.; Silva, C.; Abbott, S.; Ramirez-Medina, E.; Espinoza, N.; Borca, M.V.; Gladue, D.P.; Diaz-San Segundo, F. A Highly Effective African Swine Fever Virus Vaccine Elicits a Memory T Cell Response in Vaccinated Swine. Pathogens 2022, 11, 1438. [Google Scholar] [CrossRef] [PubMed]
  14. Goatley, L.C.; Nash, R.H.; Andrews, C.; Hargreaves, Z.; Tng, P.; Reis, A.L.; Graham, S.P.; Netherton, C.L. Cellular and Humoral Immune Responses after Immunisation with Low Virulent African Swine Fever Virus in the Large White Inbred Babraham Line and Outbred Domestic Pigs. Viruses 2022, 14, 1487. [Google Scholar] [CrossRef] [PubMed]
  15. Argilaguet, J.M.; Perez-Martin, E.; Nofrarias, M.; Gallardo, C.; Accensi, F.; Lacasta, A.; Mora, M.; Ballester, M.; Galindo-Cardiel, I.; Lopez-Soria, S.; et al. DNA vaccination partially protects against African swine fever virus lethal challenge in the absence of antibodies. PLoS ONE 2012, 7, e40942. [Google Scholar] [CrossRef] [PubMed]
  16. Bosch-Camos, L.; Lopez, E.; Navas, M.J.; Pina-Pedrero, S.; Accensi, F.; Correa-Fiz, F.; Park, C.; Carrascal, M.; Dominguez, J.; Salas, M.L.; et al. Identification of Promiscuous African Swine Fever Virus T-Cell Determinants Using a Multiple Technical Approach. Vaccines 2021, 9, 29. [Google Scholar] [CrossRef] [PubMed]
  17. Yang, J.; Li, S.; Feng, T.; Zhang, X.; Yang, F.; Cao, W.; Chen, H.; Liu, H.; Zhang, K.; Zhu, Z.; et al. African Swine Fever Virus F317L Protein Inhibits NF-kappaB Activation to Evade Host Immune Response and Promote Viral Replication. mSphere 2021, 6, e00658-21. [Google Scholar] [CrossRef] [PubMed]
  18. Kong, J.; Shao, Y.; Qiao, Y.; Zhao, Q.; Li, H.; Zhang, X.; Lin, W.; Chen, W.; Xie, Q. Using Red/ET recombination technology to construct recombinant pseudorabies virus expressing African swine fever virus CP530R and F317L genes. J. Chin. Vet. Med. 2021, 41, 1716–1721. (In Chinese) [Google Scholar] [CrossRef]
  19. Liu, S.; Luo, Y.; Wang, Y.; Li, S.; Zhao, Z.; Bi, Y.; Sun, J.; Peng, R.; Song, H.; Zhu, D.; et al. Cryo-EM Structure of the African Swine Fever Virus. Cell Host Microbe 2019, 26, 836–843.e3. [Google Scholar] [CrossRef]
  20. Tlaxca, J.L.; Ellis, S.; Remmele, R.L., Jr. Live attenuated and inactivated viral vaccine formulation and nasal delivery: Potential and challenges. Adv. Drug Deliv. Rev. 2015, 93, 56–78. [Google Scholar] [CrossRef]
  21. Ma, Y.; Shao, J.; Liu, W.; Gao, S.; Peng, D.; Miao, C.; Yang, S.; Hou, Z.; Zhou, G.; Qi, X.; et al. A vesicular stomatitis virus-based African swine fever vaccine prototype effectively induced robust immune responses in mice following a single-dose immunization. Front. Microbiol. 2023, 14, 1310333. [Google Scholar] [CrossRef] [PubMed]
  22. Cadenas-Fernandez, E.; Sanchez-Vizcaino, J.M.; van den Born, E.; Kosowska, A.; van Kilsdonk, E.; Fernandez-Pacheco, P.; Gallardo, C.; Arias, M.; Barasona, J.A. High Doses of Inactivated African Swine Fever Virus Are Safe, but Do Not Confer Protection against a Virulent Challenge. Vaccines 2021, 9, 242. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, Q.; Ma, B.; Qian, N.; Zhang, F.; Tan, X.; Lei, J.; Xiang, Y. Structure of the African swine fever virus major capsid protein p72. Cell Res. 2019, 29, 953–955. [Google Scholar] [CrossRef] [PubMed]
  24. Wozniakowski, G.; Mazur-Panasiuk, N.; Walczak, M.; Juszkiewicz, M.; Frant, M.; Niemczuk, K. Attempts at the Development of a Recombinant African Swine Fever Virus Strain with Abrogated EP402R, 9GL, and A238L Gene Structure using the CRISPR/Cas9 System. J. Vet. Res. 2020, 64, 197–205. [Google Scholar] [CrossRef] [PubMed]
  25. Gomez-Puertas, P.; Rodriguez, F.; Oviedo, J.M.; Brun, A.; Alonso, C.; Escribano, J.M. The African swine fever virus proteins p54 and p30 are involved in two distinct steps of virus attachment and both contribute to the antibody-mediated protective immune response. Virology 1998, 243, 461–471. [Google Scholar] [CrossRef] [PubMed]
  26. Barderas, M.G.; Rodriguez, F.; Gomez-Puertas, P.; Aviles, M.; Beitia, F.; Alonso, C.; Escribano, J.M. Antigenic and immunogenic properties of a chimera of two immunodominant African swine fever virus proteins. Arch. Virol. 2001, 146, 1681–1691. [Google Scholar] [CrossRef] [PubMed]
  27. Lokhandwala, S.; Waghela, S.D.; Bray, J.; Martin, C.L.; Sangewar, N.; Charendoff, C.; Shetti, R.; Ashley, C.; Chen, C.H.; Berghman, L.R.; et al. Induction of Robust Immune Responses in Swine by Using a Cocktail of Adenovirus-Vectored African Swine Fever Virus Antigens. Clin. Vaccine Immunol. 2016, 23, 888–900. [Google Scholar] [CrossRef]
  28. Argilaguet, J.M.; Perez-Martin, E.; Lopez, S.; Goethe, M.; Escribano, J.M.; Giesow, K.; Keil, G.M.; Rodriguez, F. BacMam immunization partially protects pigs against sublethal challenge with African swine fever virus. Antiviral Res. 2013, 98, 61–65. [Google Scholar] [CrossRef]
  29. Cadenas-Fernandez, E.; Sanchez-Vizcaino, J.M.; Kosowska, A.; Rivera, B.; Mayoral-Alegre, F.; Rodriguez-Bertos, A.; Yao, J.; Bray, J.; Lokhandwala, S.; Mwangi, W.; et al. Adenovirus-vectored African Swine Fever Virus Antigens Cocktail Is Not Protective against Virulent Arm07 Isolate in Eurasian Wild Boar. Pathogens 2020, 9, 171. [Google Scholar] [CrossRef]
  30. Lopera-Madrid, J.; Osorio, J.E.; He, Y.; Xiang, Z.; Adams, L.G.; Laughlin, R.C.; Mwangi, W.; Subramanya, S.; Neilan, J.; Brake, D.; et al. Safety and immunogenicity of mammalian cell derived and Modified Vaccinia Ankara vectored African swine fever subunit antigens in swine. Vet. Immunol. Immunopathol. 2017, 185, 20–33. [Google Scholar] [CrossRef]
  31. Turlewicz-Podbielska, H.; Kuriga, A.; Niemyjski, R.; Tarasiuk, G.; Pomorska-Mol, M. African Swine Fever Virus as a Difficult Opponent in the Fight for a Vaccine-Current Data. Viruses 2021, 13, 1212. [Google Scholar] [CrossRef] [PubMed]
  32. Chen, W.; Zhao, D.; He, X.; Liu, R.; Wang, Z.; Zhang, X.; Li, F.; Shan, D.; Chen, H.; Zhang, J.; et al. A seven-gene-deleted African swine fever virus is safe and effective as a live attenuated vaccine in pigs. Sci. China Life Sci. 2020, 63, 623–634. [Google Scholar] [CrossRef]
  33. Monteagudo, P.L.; Lacasta, A.; Lopez, E.; Bosch, L.; Collado, J.; Pina-Pedrero, S.; Correa-Fiz, F.; Accensi, F.; Navas, M.J.; Vidal, E.; et al. BA71DeltaCD2: A New Recombinant Live Attenuated African Swine Fever Virus with Cross-Protective Capabilities. J. Virol. 2017, 91, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, Z.; Ai, Q.; Huang, S.; Ou, Y.; Gao, Y.; Tong, T.; Fan, H. Immune Escape Mechanism and Vaccine Research Progress of African Swine Fever Virus. Vaccines 2022, 10, 344. [Google Scholar] [CrossRef] [PubMed]
  35. Qu, H.; Ge, S.; Zhang, Y.; Wu, X.; Wang, Z. A systematic review of genotypes and serogroups of African swine fever virus. Virus Genes. 2022, 58, 77–87. [Google Scholar] [CrossRef]
  36. Portugal, R. ELISpot Assay for the Detection of ASFV-Specific Interferon-Gamma (IFN-gamma)-Producing Cells. Methods Mol. Biol. 2022, 2503, 169–178. [Google Scholar] [CrossRef] [PubMed]
  37. Wong, P.; Pamer, E.G. CD8 T cell responses to infectious pathogens. Annu. Rev. Immunol. 2003, 21, 29–70. [Google Scholar] [CrossRef] [PubMed]
  38. Esparza, I.; Gonzalez, J.C.; Vinuela, E. Effect of interferon-α, interferon-γ and tumour necrosis factor on African swine fever virus replication in porcine monocytes and macrophages. J. Gen. Virol. 1988, 69 Pt 12, 2973–2980. [Google Scholar] [CrossRef] [PubMed]
  39. Canter, J.A.; Aponte, T.; Ramirez-Medina, E.; Pruitt, S.; Gladue, D.P.; Borca, M.V.; Zhu, J.J. Serum Neutralizing and Enhancing Effects on African Swine Fever Virus Infectivity in Adherent Pig PBMC. Viruses 2022, 14, 1249. [Google Scholar] [CrossRef]
  40. Barry, M.; Bleackley, R.C. Cytotoxic T lymphocytes: All roads lead to death. Nat. Rev. Immunol. 2002, 2, 401–409. [Google Scholar] [CrossRef]
  41. Gerner, W.; Kaser, T.; Saalmuller, A. Porcine T lymphocytes and NK cells--an update. Dev. Comp. Immunol. 2009, 33, 310–320. [Google Scholar] [CrossRef]
  42. Oura, C.A.L.; Denyer, M.S.; Takamatsu, H.; Parkhouse, R.M.E. In vivo depletion of CD8+ T lymphocytes abrogates protective immunity to African swine fever virus. J. Gen. Virol. 2005, 86 Pt 9, 2445–2450. [Google Scholar] [CrossRef] [PubMed]
  43. Sun, L.; Niu, J.; Zhang, J.; Peng, Y.; Feng, X.; Huang, F.; Liu, J.; Li, S.; Chen, Z. Thermostable T Cell Multiepitope Nanoparticle Antigens Inducing Potent Immune Responses against the Swine Fever Virus. ACS Infect. Dis. 2023, 9, 2358–2368. [Google Scholar] [CrossRef] [PubMed]
  44. Bosch-Camos, L.; Lopez, E.; Collado, J.; Navas, M.J.; Blanco-Fuertes, M.; Pina-Pedrero, S.; Accensi, F.; Salas, M.L.; Mundt, E.; Nikolin, V.; et al. M448R and MGF505-7R: Two African Swine Fever Virus Antigens Commonly Recognized by ASFV-Specific T-Cells and with Protective Potential. Vaccines 2021, 9, 508. [Google Scholar] [CrossRef] [PubMed]
  45. Lu, W.; Bai, Y.; Zhang, S.; Zhao, X.; Jin, J.; Zhu, X.; Wang, R.; Wu, Y.; Zhang, A.; Zhang, G.; et al. An Intracellular Epitope of ASFV CD2v Protein Elicits Humoral and Cellular Immune Responses. Animals 2023, 13, 1967. [Google Scholar] [CrossRef]
  46. Herrera, L.R.M.; Bisa, E.P. In silico analysis of highly conserved cytotoxic T-cell epitopes in the structural proteins of African swine fever virus. Vet. World 2021, 14, 2625–2633. [Google Scholar] [CrossRef]
Figure 1. Experimental procedures of the immunization and sample collection. The first immunization was performed on Day 0, with a booster immunization at Day 21, and serums were collected on Days 7, 14, 21, and 28, while splenic lymphocytes were collected on Day 28. Red dots represent serum.
Figure 1. Experimental procedures of the immunization and sample collection. The first immunization was performed on Day 0, with a booster immunization at Day 21, and serums were collected on Days 7, 14, 21, and 28, while splenic lymphocytes were collected on Day 28. Red dots represent serum.
Animals 14 01331 g001
Figure 2. The IFN-γ secreted by splenic lymphocytes of immunized mice detected by IFN-γ ELISpot assays. (A) IFN-γ ELISpot spot plots. Splenic lymphocytes stimulated with 100 ng/well of F317L protein and p72 protein. (B) Histogram of spots counted in IFN-γ ELISpot assays. Data are expressed as mean ± SD. According to the number of spots counted in IFN-γ ELISpot assays, the data are representative of the mean ± SD calculated based on three mice and three parallel holes in each mouse. The difference between the F317L group compared to the negative control group (p < 0.001) and the p72 positive control group (p < 0.01) was statistically significant. Note: ** p < 0.01; *** p < 0.001.
Figure 2. The IFN-γ secreted by splenic lymphocytes of immunized mice detected by IFN-γ ELISpot assays. (A) IFN-γ ELISpot spot plots. Splenic lymphocytes stimulated with 100 ng/well of F317L protein and p72 protein. (B) Histogram of spots counted in IFN-γ ELISpot assays. Data are expressed as mean ± SD. According to the number of spots counted in IFN-γ ELISpot assays, the data are representative of the mean ± SD calculated based on three mice and three parallel holes in each mouse. The difference between the F317L group compared to the negative control group (p < 0.001) and the p72 positive control group (p < 0.01) was statistically significant. Note: ** p < 0.01; *** p < 0.001.
Animals 14 01331 g002
Figure 3. Results of T cell subpopulation assays. (A) Flow cytometry results for each immunization group. (B) Histogram of CD4+/CD8+ ratio. Statistical analysis of the T cell subset ratios showed significant differences in the F317L protein group compared with the negative control group (p < 0.01) and the p72 positive control group (p < 0.05). Note: * p < 0.05; ** p < 0.01.
Figure 3. Results of T cell subpopulation assays. (A) Flow cytometry results for each immunization group. (B) Histogram of CD4+/CD8+ ratio. Statistical analysis of the T cell subset ratios showed significant differences in the F317L protein group compared with the negative control group (p < 0.01) and the p72 positive control group (p < 0.05). Note: * p < 0.05; ** p < 0.01.
Animals 14 01331 g003
Figure 4. Specific antibody levels in humoral immunity in immunized mice. The levels of specific antibodies in the serum of 7 d, 14 d, 21 d, and 28 d in each group were detected, and the line graphs of specific antibodies were plotted.
Figure 4. Specific antibody levels in humoral immunity in immunized mice. The levels of specific antibodies in the serum of 7 d, 14 d, 21 d, and 28 d in each group were detected, and the line graphs of specific antibodies were plotted.
Animals 14 01331 g004
Figure 5. The contents of cytokines IL2, IL4, IFN-γ, and TNF-α in the serum of mice on 21 and 28 days. Notably, 21 d represents the serum sample on the 21st day after the first immunization, while 28 d represents the serum sample on the 7th day after the second strengthened immunization. It is statistically significant. Note: *** p < 0.001.
Figure 5. The contents of cytokines IL2, IL4, IFN-γ, and TNF-α in the serum of mice on 21 and 28 days. Notably, 21 d represents the serum sample on the 21st day after the first immunization, while 28 d represents the serum sample on the 7th day after the second strengthened immunization. It is statistically significant. Note: *** p < 0.001.
Animals 14 01331 g005
Figure 6. Results of IFN-γ ELISpot experiments. (A) F317L 29 peptide, protein, negative and positive control IFN-γ ELISpot spot plots (only one mouse spot plot is shown). (B) Histogram of IFN-γ ELISpot experiment results. With positive control established, the results showed that peptide F25 (246SRRSLVNPWT255) had a statistically significantly higher number of spots per SFC/105 cells than F317L protein (p < 0.01). Note: ** p < 0.01.
Figure 6. Results of IFN-γ ELISpot experiments. (A) F317L 29 peptide, protein, negative and positive control IFN-γ ELISpot spot plots (only one mouse spot plot is shown). (B) Histogram of IFN-γ ELISpot experiment results. With positive control established, the results showed that peptide F25 (246SRRSLVNPWT255) had a statistically significantly higher number of spots per SFC/105 cells than F317L protein (p < 0.01). Note: ** p < 0.01.
Animals 14 01331 g006
Table 1. Sequence information table of predicted and synthesized protein peptides.
Table 1. Sequence information table of predicted and synthesized protein peptides.
NumberPeptideNumberPeptideNumberPeptide
F11MVETQMDKL9F11115DDDREWCGR119F21178CGGMPSICD186
F226SNAHITQTM34F12117CGRINMING125F22204IISSNQVGM212
F340ENHSVDGGA48F13123INGVPEIVE131F23211GMLTVDKRII220
F446GGAAKNVSK54F14129IVEIIPSPY137F24219IIVDLWANEN228
F553SKGKSSPKE61F15137YRAGENIYF145F25246SRRSLVNPWT255
F657SSPKEKKHW65F16148EAMMPADIY156F26262ILQDYGIEY270
F760KEKKHWTEF68F17157SRVANKPAMF166F27270YIIFPSNDF278
F863KHWTEFESW71F18160ANKPAMFVF168F28276NDFFIYEDE284
F973QLSKSKRSF81F19166FVFHTHPNL174F29294TNFFTLHEL302
F1076KSKRSFKEY84F20172PNLGSCCGG180
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

Huang, Y.; Zhai, W.; Wang, Z.; He, Y.; Tao, C.; Chu, Y.; Pang, Z.; Zhu, H.; Jia, H. Analysis of the Immunogenicity of African Swine Fever F317L Protein and Screening of T Cell Epitopes. Animals 2024, 14, 1331. https://doi.org/10.3390/ani14091331

AMA Style

Huang Y, Zhai W, Wang Z, He Y, Tao C, Chu Y, Pang Z, Zhu H, Jia H. Analysis of the Immunogenicity of African Swine Fever F317L Protein and Screening of T Cell Epitopes. Animals. 2024; 14(9):1331. https://doi.org/10.3390/ani14091331

Chicago/Turabian Style

Huang, Ying, Wenzhu Zhai, Zhen Wang, Yuheng He, Chunhao Tao, Yuanyuan Chu, Zhongbao Pang, Hongfei Zhu, and Hong Jia. 2024. "Analysis of the Immunogenicity of African Swine Fever F317L Protein and Screening of T Cell Epitopes" Animals 14, no. 9: 1331. https://doi.org/10.3390/ani14091331

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