Abstract Using nanoparticles (NPs) as a platform for multivalent antigen display is an effective strategy to increase the immunogenicity of subunit vaccines, which can induce high levels of humoral and cellular immunity. In addition, antigens that target antigen-presenting cells (APCs) can further increase their immunogenicity. To date, there are no commercially available ASFV vaccines available worldwide. The present study developed a dendritic cell (DC)-targeting ASFV biomimetic nanovaccine. First, a high-affinity and specific nanobody (Nb) targeting DCs was screened and expressed in tandem with B and T-cell epitopes of highly immunogenic p30, p54, p72, pB602L, and CD2V proteins of ASFV (Nb-rAg). The Nb-rAg complexes were then loaded onto azabisphosphonate-terminated phosphorus dendrimers (PPHs) to construct PPH-Nb-rAg NPs, which were subsequently coated with ASFV-infected activated porcine alveolar macrophage (PAM) membranes to prepare the PPH-Nb-rAg@PM biomimetic nanovaccine. Finally, the immune efficacy of the nanovaccine was evaluated in mice. Notably, compared with the PBS, rAg, Nb-rAg, and PPH-Nb-rAg immunization groups, the PPH-Nb-rAg@PM immunization group exhibited stronger ASFV antigen-specific humoral and cellular immune responses. Single-cell RNA sequencing (scRNA-seq) revealed that immunization with PPH-Nb-rAg@PM increased the proportions of B cells, T cells, NK cells, plasma cells, and macrophages in the mouse spleen. Further analysis revealed that PPH-Nb-rAg@PM immunization increased the numbers of memory B cells and plasma cells in the mouse spleen, and the numbers of CD4 + T cells, CD8 + T cells and NK cells also increased compared with those in the control group. These results suggest that PPH-Nb-rAg@PM is a promising and effective candidate vaccine against ASFV. Graphical abstract [40]graphic file with name 12951_2025_3593_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03593-7. Keywords: African swine fever, Multiepitope nanovaccine, B-cell response, T-cell response Introduction African swine fever (ASF) is caused by the ASF virus (ASFV), and its clinical syndrome is characterized by rapid onset and symptoms such as high fever, dyspnea, and extensive hemorrhaging of the skin and various internal organs in pigs [[41]1, [42]2]. Since August 2018, ASF has spread throughout China and other adjacent Asian countries, causing substantial losses to the pig industry in these regions [[43]3, [44]4]. ASFV is an enveloped double-stranded DNA virus belonging to the Asfarviridae family, and its genome is approximately 170 ~ 193 kb and encodes 150 ~ 167 proteins [[45]5]. This virus is able to evade elimination by the host immune system effectively, and viral protein functions are mostly unknown, leading to considerable obstacles in the development of ASF vaccines. Most reported vaccines still have various problems and cannot or can only partially exert protective effects. For example, some natural or artificial attenuated vaccines or attenuated gene knockout vaccines may have obvious side effects or provide protection only against specific ASFV strains, whereas inactivated vaccines cannot effectively protect experimental pigs from virulent strain infection [[46]6–[47]9]. In addition, subunit vaccines and DNA vaccines also cannot effectively protect pigs from infection [[48]10–[49]12]. There is no commercially available ASF vaccine thus far. Owing to the complexity of the structure and function of the ASFV protein, screening protective antigens suitable for application in subunit vaccine development is difficult. Nevertheless, some viral proteins can induce neutralizing antibodies and have certain viral neutralizing activities, providing the possibility for the development of new types of vaccines. The ASFV p54 and p30 proteins can induce neutralizing antibodies in pigs, but immunized pigs are not fully protected against lethal infections [[50]13]. In addition to inducing neutralizing antibodies, p72 and p30 can also activate cytotoxic T lymphocyte responses [[51]11]. Other ASFV envelope or membrane proteins, such as CD2v, p12, and D117L, may also induce neutralizing antibodies and provide serotype-specific cross-protective immunity [[52]14, [53]15]. Oura et al. confirmed that the clearance of CD8 + T cells led to complete loss of resistance in pigs to infection with the virulent ASFV OUR/T88/1 strain [[54]16], indicating that relying solely on humoral immunity cannot provide effective immune protection against ASFV infection and that cellular immunity also plays an important role in immune protection, emphasizing that T-cell epitopes are necessary for the development of ASF vaccines. Dendritic cells (DCs) serve as a bridge between innate and adaptive immunity and are critical in initiating immune responses and immune regulation [[55]17]. Immature DCs continuously capture pathogens such as viruses and bacteria to activate themselves into mature DCs and migrate to lymph nodes, where they act as antigen-presenting cells (APCs) to present antigens and nonantigen-specific costimulatory signals from pathogens to T cells to activate helper T cells, killing T cells and B cells [[56]18]. Therefore, DCs are usually utilized as antigen targets for the induction of efficient and strong immune responses. For the design of DC-targeting vaccines, the practical application of traditional DC receptor-specific monoclonal antibodies (McAbs) [[57]19–[58]23], ligands [[59]24], and single-chain variable fragments [[60]25] is greatly restricted because of their enormous molecular weight, complex structure and high cost. As an alternative, the heavy chain variable region, also known as the nanobody (Nb), is isolated from camelids, providing another complete, new and much smaller DC-targeting vehicle. Compared with traditional antibodies, Nbs have many unique advantages, such as a smaller molecular weight (~ 15 vs. 150 kDa), smaller size (~ 4 vs. 14.2 nm), high affinity, ease of modification and engineering, and low immunogenicity [[61]26, [62]27], making them ideal antigen delivery vehicles for DC targeting. XC motif chemokine receptor 1 (XCR1) is selectively expressed on CD8 + DCs with cross-antigen presentation ability [[63]28], enhances specific cytotoxic T cells [[64]29], and is highly conserved across different species, making XCR1 + DC targeting a new strategy for vaccine development. The present study aimed to screen porcine XCR1 (pXCR1)-specific Nbs, and a novel ASFV antigen delivery system based on the screened Nbs was constructed to evaluate their immune potential. Dendritic macromolecules have precise molecular structures, perfect spatial geometric symmetry, many surface functional groups and internal cavities, and controllable nanoscale and highly branched three-dimensional structures. These characteristics make dendritic macromolecules important carriers for delivering drugs, genes, and diagnostic reagents. The major advantages of these nonviral delivery systems over traditional viral vectors can be highlighted in their simplicity of synthesis, strong stability, tunable modifications, and biosafety profile [[65]30]. Azabis phosphonate-terminated phosphorus dendrimers (PPHs) with iminodi (methylphosphate) terminals can serve as nanocarriers to load drugs and have anti-inflammatory activity, which can activate the immune system and induce natural killer (NK) cell proliferation; the latter plays an important role in immune regulation in anticancer and anti-infection activities [[66]31]. In addition, PPHs with iminodi (methylphosphate) termini have been proven to be a universal protein delivery system since they possess terminal phosphate anionic groups and a benzene ring-rich branched scaffold structure and can complex proteins through electrostatic interactions, hydrogen bonding, cation-Π interactions, and hydrophobic interactions [[67]32]. As an appealing emerging cell-mimicking platform, cell membrane-coated biomimetic nanovaccines may have better performance in the face of ASFV challenge. Cell membrane-coated biomimetic nanoparticles (NPs) can perform multiple functions, such as targeted recognition and immunomodulation, which are attributed to their inherent expression of diverse pathogen-associated receptors (PRRs) on derived cell membranes [[68]33, [69]34]. Additionally, nanomedicines can be enhanced after they are camouflaged with preactivated macrophage membranes (PMs) because of their immune escape ability and prolonged circulation, which results in less clearance by the mononuclear phagocyte system [[70]35, [71]36]. For example, a PM-camouflaged nanoplatform consisting of branched polyethylenimine, short hairpin RNA targeting Ptpn2 and doxorubicin can reduce phagocytosis by macrophages and significantly prolong the circulation time in vivo, thus enhancing the antitumor activity of NPs [[72]37]. However, no biomimetic NPs have been applied for the development of ASF vaccines. Porcine alveolar macrophages (PAMs), the main target cells after ASFV infection in pigs in vivo, and one of the pivotal members, as the first line defence against invaded viruses, may abundantly express specific receptor(s) that target ASFV when infected by ASFV. ASFV infection can also activate many types of PRRs (such as RIG-I, MDA5 and LGP2) of the Toll-like receptor (TLR) pathway [[73]38], which can recognize specific pathogen-associated molecular patterns (PAMPs), activate innate immunity and exert anti-viral effects [[74]39]. Therefore, we hypothesized that ASFV-infected PAM membranes may likewise express high levels of ASFV-relevant TLRs on their membrane surface and that biomimetic NPs coated with these PMs may have longer retention times than traditional vaccines do. This study developed a DC-targeting ASFV biomimetic nanovaccine. First, ASFV B-cell epitopes that can induce neutralizing antibodies and T-cell epitopes that can induce a cellular immune response were screened and combined with a recombinant antigen (rAg); phage display technology was used to screen specific Nbs against the DC surface ligand XCR1, and the Nbs and rAg genes were tandemly expressed to prepare Nb-rAg; and Nb-rAg was coupled with PPH phosphorus dendrimers to prepare PPH-Nb-rAg NPs, which were then coated with ASFV-infected PAM membranes to prepare the PPH-Nb-rAg@PM biomimetic nanovaccine. Finally, the immune efficacy of the nanovaccine was evaluated in mice. The advantages of this new vaccine are as follows: it utilizes phosphate terminal dendritic molecules coupled with Nb-rAg targeted to DCs and disguises them with preactivated membranes to construct NPs. Simple ingredients and longer circulation times in vivo. Compared with ordinary vaccines, the different components of NPs can activate NK cell proliferation, inducing more durable, stronger cellular and humoral immune responses. Materials and methods Screening, identification and recombination of immune-dominant B and T-cell epitopes To optimize the immune characteristics of the vaccine as much as possible and improve the antibody level and activity after immunization, neutralizing active epitopes in ASFV proteins were carefully screened according to previous reports. These epitopes were distributed mainly in the p30, p54, p72, pB602L, and CD2V proteins of ASFV. These preliminarily screened epitopes were then artificially synthesized and further identified for their reactivity with inactivated ASFV-antibody-positive pig serum to determine B-cell epitopes via dot-ELISA. Briefly, the synthesized epitope peptide was dissolved in phosphate-buffered saline (PBS) and coated onto an ELISA plate (2 µg/well) at 4°C overnight. Then, the peptide coating solution was discarded, and the samples were blocked with 200 µl/well 2.5% dry milk for 2 h at 37°C. After washing with PBST 3 times, each peptide was incubated with 10 portions of inactivated pig anti-ASFV antibody-positive serum (1:10 dilution) for 1 h at 37°C. The plates were washed 3 times with PBST and then incubated with horseradish peroxidase (HRP)-conjugated goat anti-swine IgG (H&L) (1:5,000). After washing with PBST 3 times, 100 µl/well 3,3’,5,5’-tetramethylbenzidine (TMB) was added, and the mixture was incubated for 15 min in the dark. Finally, 3 M H[2]SO[4] (50 µl/well) was added to stop the colorimetric reaction, and the optical density (OD) at 450 nm was read via an automated ELISA plate reader (BioTek; Agilent Technologies, Inc., USA). In accordance with previous reports, T-cell epitope peptides were synthesized to evaluate their ability to activate splenocyte activation and subsequent IFN-γ secretion. Briefly, 6-week-old mice were immunized with ASFV p30, p72, or CD2v recombinant protein. At 7 d after immunization, the spleens were dissected, and the splenocytes were isolated, counted, and seeded into a 96-well plate at a density of 1 × 10^6/ml per well. Then, 10 µg of epitope peptide dissolved in PBS was added to each well, and the plate was cultured at 37 °C in a 5% CO[2] incubator for 60 h. Cell proliferation was measured via the Cell Counting Kit-8 (CCK-8) assay. Then, the cells were plated and treated as described above, and an ELISpot assay was performed to analyze the proportion of IFN-γ-positive splenocytes. Non-immunized healthy mouse splenocytes were used as the negative control. Preparation of pXCR1-specific Nb and Nb-rAg recombinant proteins The pXCR1 gene was synthesized, cloned, and inserted into the pET-30a expression vector. After that, the pET-30a-pXCR1-His plasmid was transformed into E. coli BL21 (DE3) competent cells, which were induced with 1.0 mM IPTG to express the pXCR1 protein. After purification with a nickel column, the pXCR1 protein was used to immunize adult male bactrian camel at a dose of 5.0 mg each time at 2-week intervals. After the 5 immunizations, the pXCR1 protein antibody titer in the camel serum was tested via indirect ELISA (iELISA). Then, whole blood was collected, peripheral blood lymphocytes (PBMCs) were isolated, and total RNA from PBMCs was extracted and reverse transcribed into cDNA, followed by amplification of the VHH gene by 2 rounds of nested PCR and cloning of the VHH gene into the phagemid display vector pCANTAB-5E through the Pst I and Not I restriction sites (NEB, Ipswich, MA, USA) and electronical transformation into fresh E. coli TG1 competent cells to construct the VHH phage display library. The M13K07 helper phage was utilized to rescue the VHH phage library, and anti-pXCR1-specific Nb(s) were screened by 4 rounds of biopanning via phage display technology. The specific phage particles were significantly enriched after 4 rounds of biopanning. Seventy-two individual colonies were randomly selected for sequencing and classification according to sequence diversity in the third complementarity-determining region (CDR3). The specificity and affinity of the Nb(s) screened were subsequently evaluated via anti-M13/HRP-conjugated phage ELISA. The gene corresponding to the Nb with the highest affinity for the pXCR1 protein was coupled with the optimized ASFV rAg gene by a flexible linker (hereafter abbreviated as Nb-rAg), and the Nb-rAg gene fragment was cloned and inserted into the pET-30a plasmid and transformed into E. coli BL 21(DE3) competent cells to induce the expression of the Nb-rAg protein. The Nb-rAg protein was purified with a nickel column for subsequent experiments. Preparation of iminodi(methylphosphate)-capped phosphorus dendrimers (PPHs) and PPH-Nb-rAg In accordance with previous reports, third-generation iminodi (methylphosphate) terminal dendrimer molecules were synthesized [[75]40]. The PPHs and Nb-rAg proteins were resolved in sterile water separately, and then the PPH was added dropwise into the Nb-rAg solution at mass ratios of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, and 8:1 to determine the optimal mass ratio of the two. The solutions were stirred at room temperature (RT) for 24 h until they turned light white. The mixtures were subsequently centrifuged at 15,000 × g at RT for 20 min to separate the solid and liquid phases, and the supernatant and precipitate were harvested separately. The concentration of residual Nb-rAg in the supernatant was assayed via a bicinchoninic acid (BCA) assay kit to calculate the encapsulation efficiency (EE) and loading capacity (LC) of the PPH-Nb-rAg NPs. The precipitate was subsequently resuspended in sterile water and stored at 4 °C until further use. The EE of Nb-rAg by the PPH-Nb-rAg NPs was calculated via the following formula: EE (%) = (initial Nb-rAg amount - unencapsulated free Nb-rAg)/initial Nb-rAg amount. The LC of Nb-rAg by the PPH-Nb-rAg NPs was calculated via the following formula: LC (%) = (initial Nb-rAg amount - unencapsulated free Nb-rAg)/total amount of PPH-Nb-rAg NPs. Preparation of the PPH-Nb-rAg@PM NPs The ASFV (0.01 MOI) and PAMs were cocultured at 37 °C for 12 h. After washing 3 times with PBS, 2 ml of ice-cold Tris-MgCl[2] buffer (pH 7.4, 0.01 M Tris, 0.001 M MgCl[2]) supplemented with 1% PMSF (v/v, to prevent protein degradation) was added, and the cells were collected via a cell scraper. An ultrasonic disruptor was used for ultrasonic lysis to obtain a cell homogenate. Then, 1 M sucrose solution was added to the cell homogenate to achieve a final concentration of 0.25 M, followed by centrifugation at 2,000 × g for 10 min at 4 °C. The supernatant was collected and centrifuged at 14,000 × g for 30 min at 4 °C, and the milky precipitates were collected. The final precipitates were PMs, which were stored at 4 °C for further use. The PPH-Nb-rAg NPs were mixed with PMs at a mass ratio of 1:1 and then coextruded through a 200 nm porous polycarbonate membrane for 20 cycles to form biomimetic PPH-Nb-rAg@PM NPs. PPH-Nb-rAg@M NPs were prepared simultaneously, where M or PM denoted the ordinary or preactivated membrane. Characterization of the membrane-encapsulated biomimetic PPH-Nb-rAg@PM NPs The size distributions and surface potentials of the PPH-Nb-rAg@M and PPH-Nb-rAg@PM NPs were analyzed via dynamic light scattering (DLS) with Malvern Zetasizer Nano ZS ZEN3600 equipment (Malvern, Worcestershire, U.K.). The morphology of the prepared PPH-Nb-rAg NPs was observed via field-emission scanning electron microscopy (FE-SEM, SU8100, Hitachi, Tokyo, Japan) at a voltage of 3.0 kV and transmission electron microscopy (TEM, Hitachi H7800, Tokyo, Japan) at an accelerating voltage of 120.0 kV. The morphology of the prepared PPH-Nb-rAg@PM NPs was also observed via TEM. For TEM imaging, the PPH-Nb-rAg, PPH-Nb-rAg@M or PPH-Nb-rAg@PM NPs in an aqueous solution (2 mg/ml, 5 µl) were deposited onto a carbon-coated copper grid and air-dried before measurement. The particle size distribution was analyzed with ImageJ software ([76]http://rsb.info.nih.gov/ij/download.html). For size distribution analysis, at least 200 particles were randomly selected from different SEM images to obtain the size distribution histogram. For the stability analysis, the PPH-Nb-rAg@PM NPs were resuspended in 10% FBS + DMEM or saline at a final concentration of 1.0 mg/ml. The particle size was measured every 3 d for 15 d, and each measurement was repeated three times. The protein components of the PPH, Nb-rAg, PPH-Nb-rAg, PPH-Nb-rAg@M NPs, PPH-Nb-rAg@PM NPs and preactivated macrophage lysates were determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis. Different samples with equal amounts of protein (30 µg/sample) were assayed via a BCA assay kit (except for the PPH that did not carry proteins), mixed with 5 × SDS loading buffer and boiled for 5 min at 95 °C. The samples were subsequently analyzed via SDS-PAGE. The expression of TLR2, TLR4, and TLR6 in the PPH, PPH-Nb-rAg@M, and PPH-Nb-rAg@PM NP groups was detected via western blotting. In vitro cytotoxicity and cellular uptake assay To evaluate the cytotoxicity of the PPH-Nb-rAg@PM NPs, mouse monocyte macrophage leukemia cells (RAW264.7) and mouse dendritic cells (DC2.4) were plated in 96-well plates at a density of 1 × 10^4 cells/well. Approximately 24 h later, the cells were incubated with 100 µl of DMEM containing 0, 30, 60, 120, or 240 µg/ml rAg, PPH, Nb-rAg, PPH-Nb-rAg, or PPH-Nb-rAg@PM NPs for 24 h. Then, cell viability was determined via a CCK-8 kit (Beyotime Biotechnology, Shanghai, China) following the manufacturer’s instructions. The cell viability was calculated on the basis of the OD values at 450 nm. For each sample, 5 parallel wells were treated and analyzed to calculate the mean ± SD. To evaluate the cellular uptake efficiency of the PPH-Nb-rAg@PM NPs by the main APCs, RAW264.7 and DC2.4 cells were seeded into 24-well plates at a density of 5 × 10^4 cells/well. Approximately 24 h later, the cells were incubated with 50 µg of rAg, Nb-rAg, PPH-Nb-rAg, or PPH-Nb-rAg@PM NPs for another 24 h. Then, the old culture medium was discarded, and the cells were fixed at -20 °C in precooled 70% alcohol at 4°C for 30 min. After washing with PBS 3 times, the cells were incubated with inactivated ASFV-antibody-positive pig serum at a dilution of 1:400 for 1 h at RT, followed by staining with Alexa Fluor 488-conjugated goat anti-swine IgG (H&L) at a dilution of 1:500 for 1 h at RT in the dark. The cell nuclei were stained with DAPI (Beyotime Biotechnology) for 5 min. Images were acquired via fluorescence microscopy (Olympus Corporation, Tokyo, Japan). In vivo biodistribution of Cy5.5-labeled PPH-Nb-rAg@PM NPs rAg, Nb-rAg, PPH-Nb-rAg, and PPH-Nb-rAg@PM NPs were labeled with the fluorescent probe Cy5.5. Six-week-old female BALB/c mice were randomly divided into 4 groups, with 3 mice in each group. Two hundred microliters of PBS containing Cy5.5-rAg, Cy5.5-Nb-rAg, PPH-Cy5.5-Nb-rAg, or PPH-Cy5.5-Nb-rAg@PM was injected through the tail vein (injection dosage: Cy5.5-rAg = 1.5 mg/kg). In vivo fluorescence imaging of the mice was performed at 4, 8, 12, 24 and 48 h postinjection via a Live Imaging System IVIS Spectrum CT (PerkinElmer, Waltham, USA) with an excitation filter at 625 nm and an emission filter at 680 nm. The mice were sacrificed at 48 h postinjection, the liver, spleen, lung, kidney and lymph nodes of each group were isolated separately, and the fluorescence distribution in each organ was detected via an in vivo imaging system. Mouse immunization Six-week-old female BALB/c mice were randomly divided into 5 groups (n = 8 for each group) and immunized subcutaneously with 200 µl of PBS containing rAg, Nb-rAg, PPH-Nb-rAg or PPH-Nb-rAg@PM NPs on d 0 and 14 (rAg = 30 µg/mouse), and aluminum hydroxide and CPG-1826 were used as adjuvants. The control group received 200 µl of PBS. Three d prior to immunization, whole blood samples were collected to separate the serum as the negative control. Then, whole blood was collected from the tail vein at 28, 35, and 49 d after immunization, and the serum was isolated and stored at -80 °C for further analysis. At 28 d after immunization, some of the mice were euthanized with carbon dioxide in an appropriate euthanasia chamber. Spleens from mice in different immunization groups were collected under sterile conditions, and splenocytes were isolated for further experiments. Serum IgG and its subtype or isotype antibody titer detection iELISA was employed to detect serum-specific IgG and its subtype or isotype antibody titers against ASFV rAg. Briefly, 400 ng/well ASFV rAg dissolved in PBS was coated in 96-well ELISA plates at 4 °C overnight. The plates were subsequently blocked with 2.5% dry milk in PBS for 2 h at RT. After being washed with PBST 3 times, the samples were incubated with serially diluted immunized mouse serum (100 µl/well) for 1 h at 37 °C, followed by washing with PBST 3 times. Then, the samples were incubated with goat anti-mouse IgG (H&L), IgG1 (H&L), IgG2a (H&L), IgG2b (H&L), IgG2c (H&L), IgG3 (H&L), IgA or IgM antibodies (Proteintech, Wuhan, China) for 1 h at 37 °C and washed with PBST 3 times. The following operations were performed as described previously for Dot-ELISA. Flow cytometry (FCM) assay At 28 d postimmunization, the spleens of the mice in each group were isolated, and splenocyte suspensions were prepared. Before the experiments, the cells were incubated with 15 g/L EDTANa[2] solution for 5 min at 37 °C to prevent red blood cell contamination. The cells were stained with a fluorescence-labeled antibody against surface marker proteins and then subjected to FCM. For the NK cell proliferation assay, the splenocytes were stained with anti-CD3-APC and anti-CD49-FITC antibodies in the dark. For T-cell immune response analysis, the cells were stained with anti-CD3-APC and anti-CD4-FITC or anti-CD8-FITC antibodies. For DC maturation detection, the cells were stained with anti-CD11c-FITC, anti-CD80-APC, anti-CD86-APC, or anti-MHCII-APC antibodies. For analysis of the GC-B-cell population, the cells were stained with anti-CD19-FITC, anti-CD95-APC, and anti-GL7-Pacific blue antibodies. For analysis of plasma cells, anti-B220-FITC and anti-CD138-APC antibodies were used. For analysis of T follicular helper (Tfh) cells, anti-CD4-FITC, anti-PD-1-APC, and anti-CXCR5-PE antibodies were used. For analysis of effector memory T cells (T[EM]) and central memory T cells (T[CM]), anti-CD4-FITC, anti-CD44-PE, and anti-CD62L-APC antibodies were used. For analysis of memory B cells, anti-B220-PE, anti-CD38-FITC, anti-IgD-PE/Cy7, anti-GL7-Pacific Blue, and anti-IgG-APC antibodies were used. The single-cell suspensions were detected via CytExpert (Beckman Coulter, USA), and the data were analyzed via CytExpert software version 2.6. ELISpot assay Antigen-specific splenocytes from immunized BALB/c mice were analyzed via mouse IFN-γ and IL-4 ELIspot kits (DAKEWE, Beijing, China). Briefly, splenocytes were isolated from each immunized group at 28 d postimmunization, and single-cell suspensions were prepared. Then, the cells were plated into 96-well commercial ELISpot plates at a density of 5 × 10^5 cells/well, and the plates were precoated with anti-IFN-γ or -IL-4 antibodies. Approximately 24 h later, the cells were restimulated with rAg at a final concentration of 10 µg/ml for 36 h, with 3 replicates for each sample. The plates were then cultured at 37 °C in 5% CO[2] for 48 h. After incubation, the plates were incubated with a biotinylated IFN-γ detection antibody and a streptavidin-HRP conjugate. 3-Amino-9-ethylcarbazole (AEC) substrate solutions were added and incubated for 20 min, followed by rinsing with ddH[2]O and drying. The number of IFN-γ- or IL-4-positive T cells was calculated via a CTL-ImmunoSpot^®S6 cell reader/ImmunoSpot 7.0.15.1 software (Cellular Technology Limited, Shaker Heights, OH, USA) and normalized to the number of spots/10^6 splenocytes. Lymphocyte proliferation assay At 28 d after immunization, spleen lymphocytes from the mice in each immunized group were collected and plated in 96-well plates at a density of 1 × 10^6 cells/ml (100 µl/well). After 24 h, the cells were restimulated with ASFV rAg at a final concentration of 10 µg/ml for 72 h at 37 °C in 5% CO[2]. Then, 10 µl of CCK-8 was added to each well, and the mixture was cultured at 37 °C in 5% CO[2] for 3 h. The proliferation of lymphocytes was evaluated via a CCK-8 assay. The cell proliferation rate was represented as the stimulation index (SI). SI= (the mean of OD 450 nm values of the immunized lymphocyte wells)/(the mean of OD 450 nm values of unimmunized cells). Statistical analysis To ensure that the results were reproducible, all the experiments were performed independently at least three times. Statistical significance was determined by Student’s t-test when two groups were compared or by one-way analysis of variance (ANOVA) when more than two groups were compared. A P-value of < 0.05 was considered statistically significant. Results Screening and identification of Nbs against pXCR1 To develop an ASF nanovaccine targeting DCs, pXCR1-specific Nb(s) were screened. The present study first expressed and purified the pXCR1 protein in prokaryotic cells. After the protein concentration was measured and immunized bactrian camel, whole blood was collected, and PBMCs were isolated. Total RNA was extracted and reverse transcribed into cDNA. Then, pXCR1-specific Nb(s) were screened via phage display technology (Fig. [77]1A). SDS-PAGE analysis revealed that the purified recombinant His-tagged pXCR1 protein was successfully obtained with the expected size of 38 kDa (Fig. [78]1B). The western blotting results indicated that the recombinant pXCR1 protein could specifically react with the anti-His McAb (Fig. [79]1C). The purified pXCR1 protein was used to immunize bactrian camel and as a coating antigen to screen specific Nbs. Fig. 1. [80]Fig. 1 [81]Open in a new tab Screening and identification of nanobodies against pXCR1 protein. (A) Schematic diagram of antigen-specific nanobody screening. (B) SDS-PAGE analysis of the purification of pXCR1 protein. (C) Western blotting identification of His-pXCR1 protein using mouse anti-His antibody as the primary antibody. (D) After 5 immunization, the serum antibody titers against pXCR1 protein. (E) Enrichment of the specific VHH phage particles against pXCR1 protein after 4 rounds of bio-panning. (F and G) ELISA analysis of periplasmic extracts reacted with pXCR1 protein from 72 clones. (H) Amino acid sequence alignment of isolated Nbs. CDR3 variable domain was used to distinguish different Nbs. (I) ELISA was performed to detect the affinity of Nb6, Nb21 and Nb50 to pXCR1 protein. (J) ELISA analysis of the reaction specificity of Nb6, Nb21 and Nb50 with pXCR1. (K) Co-ip identification of Nb21 interacted with pXCR1 protein, Nb35 was used as a negative control. (L) Confocal analysis of screened Nb21 reactivity with pXCR1 protein. Images were obtained using a Zeiss LSM510 laser scanning inverted confocal microscope with a Zeiss 63X/1.4 NA oil immersion objective. Scale bar: 10 μm The anti-serum titer of bactrian camel against the pXCR1 protein was 1:256,000 after 5 immunizations according to the iELISA results (Fig. [82]1D), suggesting a good immune response of bactrian camel to the pXCR1 protein. Then, PBMCs were isolated from whole blood to extract total RNA, which was reverse transcribed into cDNA. After nested PCR amplification, double digestion, ligation, and transformation, a phage display VHH library was successfully constructed against the pXCR1 protein, consisting of approximately 4.2 × 10^8 individual colonies with good diversity (data not shown). After 4 rounds of biopanning, the specific VHH phage particles against the pXCR1 protein were enriched (Fig. [83]1E). Then, 72 individual colonies were randomly selected from the products of the fourth round of biopanning, and periplasmic extracts were extracted for further iELISA. Among the 72 individual colonies, 41 specifically bind with the pXCR1 protein (Fig. [84]1F and G) and were subsequently sequenced. Finally, 3 different pXCR1-specific Nbs were successfully screened according to the amino acid sequence of the CDR3 hypervariable region and named pXCR1-Nb6, -Nb21, and -Nb50 (Fig. [85]1H). pXCR1-Nb21 showed the highest affinity (Fig. [86]1I) and good specificity (Fig. [87]1J) for the pXCR1 protein. Coimmunoprecipitation (co-IP) was performed to detect whether the prepared Nb21 could react with pXCR1. Co-IP revealed that Nb21, but not the control Nb35 against the ASFV p54 protein screened previously, pulled down pXCR1 (Fig. [88]1K). Confocal microscopy revealed that Nb21 effectively reacted with XCR1 in DC2.4 cells (Fig. [89]1L). Therefore, Nb21 was selected as the delivery vehicle for ASFV antigens in the following experiments. Screening and identification of ASFV dominant B- and T-cell epitopes 41 B-cell epitopes and 11 T-cell epitopes of the p30, p54, p72, CD2v, and pB602L proteins were identified on the basis of previous studies in the NCBI database (Tables [90]S1 and [91]S2). To further screen the dominant B-cell epitopes used for the development of the ASFV candidate vaccine, these epitope peptides were synthesized and used as coated antigens, and inactivated positive sera from ASFV-recovered pigs were used as primary antibodies to perform Dot-ELISAs to detect their reactivity. The results revealed that the p30-B1, -B6, p54-B1, -B6, p72-B10, -B13, CD2v-B1, -B2, -B6, and pB602L-B3, -B7 peptides reacted well with the positive serum of ASFV-recovered pigs (OD450[nm] > 1.5) (Additional file 1: Figure [92]S1), and they were selected as the dominant B-cell epitopes for nanovaccine design. For T-cell epitopes, the identified T-cell epitope peptides were synthesized, and their ability to promote splenocyte proliferation was first tested. When the threshold was set to a cell proliferation rate greater than 30%, p30-T2, p72-T3, and CD2V-T2 cells met the requirements (Additional file 2: Figure [93]S2A). ELISpot analysis of T-cell epitopes stimulating IFN-γ secretion also revealed that p30-T2, p72-T3, and CD2V-T2 exhibited stronger stimulating effects (Additional file 2: Figure [94]S2B). Thus, these epitopes were selected as the dominant T-cell epitopes for ASFV candidate vaccine design. These selected epitopes are highly conserved between different type II ASFV strains (Additional file 3: Figure [95]S3) and are currently widely prevalent in China and Southeast Asian countries. In addition, the molecular simulation results revealed that these epitopes are distributed on the surface of the corresponding protein structure (Additional file 4: Figure [96]S4). Transcriptome analysis of ASFV-infected PAM membranes To obtain ASFV-infected PAM membranes, ASFV was cocultured with PAMs for 12 h. Then, the changes in the gene transcriptome of ASFV-infected PAMs were analyzed via high-throughput transcriptome sequencing technology. The heatmap revealed that the host genes whose expression was upregulated after ASFV infection were strongly associated with chemokines, proinflammatory cytokines, and the antiviral immune response (Additional file 5: Figure [97]S5A). Gene Ontology (GO) analysis revealed that ASFV stimulation activated the immune response, pattern recognition receptors (PRRs), inflammatory response, and inflammation-related pathways (Additional file 5: Figure [98]S5B). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that ASFV stimulation led to enrichment of the TLR signaling pathway, inflammation-related pathways and PRRs (Additional file 5: Figure [99]S5C). As the main PRRs expressed on the surface of macrophages and other immune cells, TLRs can recognize different PAMPs derived from exogenous bacteria, viruses, parasites, etc [[100]41]. Thus, TLR expression in PAMs infected with ASFV was detected via qPCR. qPCR revealed that the expression of TLR2, TLR4, and TLR6 mRNAs gradually increased with increasing stimulation time (Additional file 5: Figure [101]S5D-F). Western blotting revealed that the expression of TLR2, TLR4 and TLR6 significantly increased (Additional file 5: Figure [102]S5G). An indirect fluorescence assay (IFA) also revealed that the expression of TLR2, TLR4 and TLR6 on both the surface and cytoplasm of PAMs also increased (Additional file 5: Figure [103]S5H). Preparation and characterization of biomimetic PPH-Nb-rAg@PM NPs The third generation of azabisphosphonate-terminated sodium phosphite terminal dendrimers (PPHs) was synthesized via a divergent iterative method with hexachlorocyclotriphosphazene as the core on the basis of previous reports (Fig. [104]2A, Additional file 6: Figure [105]S6) [[106]40]. Then, prokaryotic expression, purification, and identification of the Nb-rAg protein were conducted. As shown in Fig. [107]2B, the purified Nb-rAg protein (~ 77 kDa) was successfully obtained, and it reacted well with inactivated ASFV-antibody-positive pig serum (Fig. [108]2C and D). Fig. 2. [109]Fig. 2 [110]Open in a new tab Characterization of PPH-Nb-rAg@PM NPs. (A) Chemical structure of the synthesized phosphorus dendrimers. (B) SDS-PAGE analysis of purified Nb-rAg protein. (C) ELISA analysis of the reactivity of rAg protein with 10 inactivated ASFV antibody-positive pig serums. (D) Western blotting analysis of rAg reactivity with inactivated ASFV antibody-positive pig serum. (E) Hydrodynamic size PPH, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs (n = 3). (F) Zeta potential of PPH, Nb-rAg, PM, PPH-Nb-rAg, PPH-Nb-rAg@M and PPH-Nb-rAg@PM NPs (n = 3). (G) Representvie SEM image of PPH-Nb-rAg. Scale bar: 500 nM. (H) Size distribution of PPH-Nb-rAg NPs. (I) Representvie TEM image of PPH-Nb-rAg NPs. Scale bar: 200 nM. (J) Representvie TEM image of PPH-Nb-rAg@M NPs. Scale bar: 200 nM. (K) Representvie TEM image of PPH-Nb-rAg@M NPs. Scale bar: 200 nM. (L) SDS-PAGE analysis of the protein composition of the prepared PPH-Nb-rAg@M, PPH-Nb-rAg@PM NPs and pre-activated macrophage cell lysates. (M) Western blotting analysis of TLR2, TLR4, TLR6 expression on PPH-Nb-rAg@M, PPH-Nb-rAg@PM NPs. (N) DLS detection of size changes of PPH-Nb-rAg@PM NPs in PBS, 10% FBS+DMEM and 0.9% saline (n = 3) In preliminary experiments, the optimal protein encapsulation efficiency (EE) and loading content (LC) of PPH were determined by using different mass ratios of PPH and Nb-rAg. When the mass ratio of PPH to Nb-rAg was 3:1 in sterile water for 24 h, the optimal EE (76.79%) and LC (34.28%) were obtained, as detected with a BCA assay kit (Table [111]S3). The hydrodynamic size of the PPH-Nb-rAg NPs was 169.0 ± 5.6 mm, which was larger than that of the PPH and Nb-rAg alone, indicating the successful formation of the PPH-Nb-rAg NPs (Fig. [112]2E). Finally, the PMs were extracted and coated on the surface of PPH-Nb-rAg via coextrusion to prepare PPH-Nb-rAg@PM NPs [[113]42]. Moreover, PPH-Nb-rAg coated with ordinary macrophage membranes (PPH-Nb-rAg@M) was also prepared and used as a control. Dynamic light scattering (DLS) analysis revealed that the hydrodynamic diameter ranges of PPH, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM were 160.8 ± 3.1, 10.8 ± 1.3, 169.0 ± 5.6, and 182.6 ± 3.7 nm, respectively (Fig. [114]2E). Compared with that of PPH-Nb-rAg, the hydrodynamic diameter of PPH-Nb-rAg@PM increased by approximately 13.0 nm, indicating the successful decoration of the surface of the PPH-Nb-rAg NPs with macrophage membranes (~ 13 nm in thickness) (Fig. [115]2E). After Ms or PMs decoration, the zeta potential changed from − 26.2 (PPH-Nb-rAg) to -11.6 mV (PPH-Nb-rAg@M) and − 12.02 mV (PPH-Nb-rAg@PM) (Fig. [116]2F), which was similar to the surface potential of the PMs. Scanning electron microscopy (SEM) revealed that the PPH-Nb-rAg NPs had a uniform spherical shape with a mean particle diameter of 169.0 nm (Fig. [117]2G and H). The TEM images revealed a typical sphere-like core-shell structure with a thin uniform membrane on the surface of both the PPH-Nb-rAg@M and PPH-Nb-rAg@PM NPs compared with the naked spherical PPH-Nb-rAg core (Fig. [118]2I-K). These results suggest that PMs were successfully coated on the surface of the PPH-Nb-rAg NPs. Next, SDS-PAGE combined with western blotting was performed to confirm the success of Nb-rAg loading and PMs decoration. SDS-PAGE analysis revealed that the protein compositions of PPH-Nb-rAg@M and PPH-Nb-rAg@PM were different from those of normal macrophage lysates (Fig. [119]2L), and a marked target band of approximately 77 kDa could be observed in the PPH-Nb-rAg@M and PPH-Nb-rAg@PM lanes. Western blotting confirmed that both the PPH-Nb-rAg@M and PPH-Nb-rAg@PM NPs carried membrane surface marker proteins, such as TLR2, TLR4, and TLR6, and their expression levels were greater in the PPH-Nb-rAg@PM than in the PPH-Nb-rAg@M NPs (Fig. [120]2M), confirming the successful loading of Nb-rAg, Ms and PMs, respectively. Next, the stability of the PPH-Nb-rAg@PM NPs at RT was determined by suspending them in 0.9% saline, PBS or 10% FBS + DMEM for 15 d. As shown in Fig. [121]2N, the hydrodynamic size of PPH-Nb-rAg@PM remained almost unchanged during incubation, indicating that the prepared PPH-Nb-rAg@PM NPs are stable. Cytotoxicity, cellular uptake and biodistribution assays A CCK-8 assay was used to evaluate the viability of RAW264.7 and DC2.4 cells after incubation with rAg, PPH, Nb-rAg, PPH-Nb-rAg or PPH-Nb-rAg@PM NPs for 24 h in vitro. The results indicated that even when the highest concentration reached 240 µg/ml, these NPs did not exhibit notable cytotoxicity to either RAW264.7 or DC2.4 cells (Fig. [122]3A and B), suggesting that the PPH-Nb-rAg@PM NPs have good cytocompatibility. Fig. 3. [123]Fig. 3 [124]Open in a new tab Cytotoxicity and in vivo distribution of prepared PPH-Nb-rAg@PM NPs. Cell viability of RAW264.7 (A) and DC2.4 (B) cells after incubation with 0, 30, 60, 120, 240 μg/ml rAg, Nb-rAg, PPH, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs for 24 h. Then cell viability of each group was detected using a CCK-8 kit. (C) Uptake of rAg, Nb-rAg, PPH-Nb-rAg, and PPH-Nb-rAg@PM NPs by RAW264.7 cells. (D) FCM analysis of fluorescence intensity of (C), the fluorescence intensity of the rAg treatment group was normalized as 100%. (E) Uptake of rAg, Nb-rAg, PPH-Nb-rAg, and PPH-Nb-rAg@PM NPs by DC2.4 cells. (F) FCM analysis of fluorescence intensity of (E), the fluorescence intensity of the rAg treatment group was normalized as 100%. (G) Detection and biodistribution of Cy5.5-rAg, -Nb-rAg, -PPH-Nb-rAg and -PPH-Nb-rAg@PM NPs. (H) Biodistribution of fluorescent antigens in the liver, spleen, lung, kidney, and lymph nodes were analyzed APCs are responsible for the uptake and processing of various antigens and play a critical role in inducing immune response. Thus, the cellular uptake of rAg, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs by RAW264.7 and DC2.4 cells was investigated via IFA. The results revealed that the fluorescence intensity of RAW264.7 cells treated with Nb-rAg was 2.47 times stronger than that of those treated with rAg, and that of the PPH-Nb-rAg group was approximately 1.77 times stronger than that of the Nb-rAg group (Fig. [125]3C and D), indicating that the conjugation of Nb-rAg with PPH promoted the cellular uptake of Nb-rAg. The fluorescence intensity of the PPH-Nb-rAg treatment group was 4.8 times stronger than that of the PPH-Nb-rAg@PM group (Fig. [126]3C and D), implying that PPH-Nb-rAg NPs decorated with PMs effectively hindered their uptake by macrophages and prolonged their half-life. For DC2.4 cells, the fluorescence intensity of the PPH-Nb-rAg@PM treatment group was 1.17 times that of the PPH-Nb-rAg group (Fig. [127]3E and F), indicating that llike PPH-Nb-rAg, PPH-Nb-rAg@PM NPs are also able to be efficiently uptaken by DC2.4 cells. To investigate the distribution of the PPH-Nb-rAg@PM NPs in vivo, BALB/c mice were intravenously injected with Cy5.5-rAg, Cy5.5-Nb-rAg, PPH-Cy5.5-Nb-rAg, or PPH-Cy5.5-Nb-rAg@PM NPs, and fluorescence imaging was performed at the indicated time points via a live imaging system. As shown in Fig. [128]3G, the fluorescence intensity of the Cy5.5-rAg group was the weakest and reached the highest value at 12 h post-injection, after which it gradually decreased. The fluorescence intensities of the Cy5.5-Nb-rAg and PPH-Cy5.5-Nb-rAg groups were both greater than those of the Cy5.5-rAg group and decreased beginning at 24 h post-injection. Notably, the fluorescence intensity of the PPH-Cy5.5-Nb-rAg@PM group was greater than that of the other three groups, and strong fluorescence continued to be observed even at 48 h post-injection (Fig. [129]3G). To further investigate the in vivo distribution of these NPs, the mice were sacrificed at 48 h, and their major organs were collected and imaged. As shown in Fig. [130]3H, the PPH-Cy5.5-Nb-rAg@PM group presented the strongest Cy5.5 fluorescence intensity in the lungs, lymph nodes, and spleens among all the groups, which was consistent with the in vivo fluorescence results, indicating that avoiding engulfment by macrophages effectively prolonged the half-life of the PPH-Nb-rAg@PM NPs in vivo. PPH-Nb-rAg@PM NPs promote NK cell proliferation Studies have shown that the PPH can induce NK cell proliferation [[131]31]. NK cells act as regulatory cells of the immune system and are able to release perforin and IFN-γ to combat virus-infected cells and to regulate innate and adaptive immune reactions. Therefore, whether PPH-Nb-rAg@PM NPs can regulate the proliferation of NK cells in mice was investigated. Mouse PBMCs were isolated and stimulated with PBS, IL-2, IL-2 + PPH, IL-2 + PPH-Nb-rAg, or IL-2 + PPH-Nb-rAg@PM, after which an FCM assay was carried out to detect the population of CD3-CD49 + phenotype NK cells. IL-2 was included as the positive control. The results revealed that the proportion of CD3-CD49 + NK cells in the IL-2 + PPH-stimulated group was 21.2%, which was 1.19 times greater than that in the IL-2 treatment group (Fig. [132]4A and B), and the proportions of CD3-CD49 + NK cells in the IL-2 + PPH-Nb-rAg and IL-2 + PPH-Nb-rAg@PM groups were 24.31% and 26.41%, respectively, which were 1.15 and 1.25 times greater than those in the IL-2 treatment group (Fig. [133]4A and B). However, the number of CD3-CD49 + NK cells in the IL-2 + PPH-Nb-rAg group and the IL-2 + PPH-Nb-rAg@PM group did not significantly differ (P > 0.05, Fig. [134]4B), indicating that the proliferation of NK cells is largely dependent on the PPH, which can inherently stimulate NK cell activation. Fig. 4. [135]Fig. 4 [136]Open in a new tab PPH-Nb-rAg@PM NPs promotes NK cells proliferation in vitro and in vivo. Representative flow cytometry plots (A) and quantification (B) of CD3-CD49 + phenotype NK cells proportion after mice PBMCs were stimulated with PBS, IL-2, IL-2 + PPH, IL-2 + PPH-Nb-rAg and IL-2 + PPH-Nb-rAg@PM NPs (n = 3). ELISA analysis of the cell culture supernatants IFN-γ (C) or perforin (D) secretion after mice PBMCs were stimulated with the above mentioned NPs (n = 3). (E and F) Represent flow cytometry plots and quantification of CD3-CD49 + NK cells in spleen of each group at 28 d after immunization (n = 3). (G) ELISA analysis of mice serum IFN-γ contents at 28 d post immunization (n = 3). (H) ELISA analysis of mice serum perforin contents at 28 d post immunization (n = 3) The expression levels of IFN-γ and perforin, two important pathogenic-killing cytokines in PBMC culture supernatants, were detected via ELISA. The ELISA results revealed that, compared with the IL-2 treatment, the IL-2 + PPH treatment dramatically stimulated the secretion of IFN-γ into the supernatant, which was increased by 1.2-fold (Fig. [137]4C), and the IL-2 + PPH-Nb-rAg and IL-2 + PPH-Nb-rAg@PM treatment groups were increased by 1.39- and 1.45-fold, respectively, compared with the IL-2 treatment group (Fig. [138]4C). There was no marked difference between the IL-2 + PPH-Nb-rAg and the IL-2 + PPH-Nb-rAg@PM groups (Fig. [139]4C). In terms of perforin, the levels in the IL-2 + PPH, IL-2 + PPH-Nb-rAg and IL-2 + PPH-Nb-rAg@PM groups were 2.04, 2.75, and 2.82 times greater than those in the IL-2 treatment groups (21.71, 29.24, 30.04 and 10.64 pg/ml, respectively), and no marked difference was observed between the IL-2 + PPH-Nb-rAg and IL-2 + PPH-Nb-rAg@PM groups (Fig. [140]4D). Next, the proliferation of NK cells in vivo was further investigated. Splenocytes were isolated from different immunized mice at 28 d after immunization and then subjected to FCM. Compared with the PBS group, immunization with rAg, Nb-rAg, or PPH-Nb-rAg or PPH-Nb-rAg@PM NPs increased the number of NK cells with the CD3-CD49 + phenotype by 1.07, 1.26, 1.37 and 1.48 times, respectively, and the PPH-Nb-rAg@PM NPs induced the highest percentage of CD3-CD49 + phenotype NK cells among all the groups (Fig. [141]4E and F). Splenocyte culture supernatant IFN-γ analysis via ELISA revealed that the PPH-Nb-rAg@PM immunization group presented the highest level of supernatant IFN-γ (Fig. [142]4G), and perforin analysis revealed similar results (Fig. [143]4H). Taken together, these results suggest that PPH decoration endows ASFV antigens with an inherent immunomodulatory ability to promote NK cell proliferation in vitro or in vivo. PPH-Nb-rAg@PM NP immunization induced humoral immune response At 28, 35, and 49 d after immunization, the levels of mouse serum antibodies against ASFV rAg were detected via ELISA with rAg as the coating antigen (Fig. [144]5A). At 28 d post-immunization, the antibody titer in the Nb-rAg immunization group was 5.35 times greater than that in the rAg group (Fig. [145]5B). Compared with the Nb-rAg group, the PPH-Nb-rAg group presented 13.4 times greater antibody titers (Fig. [146]5B). The greatest specific antibody titers were induced via immunization with PPH-Nb-rAg@PM NPs, which was 55.2 times greater than that of the PPH-Nb-rAg group (Fig. [147]5B). ELISA analysis of the serum antibody titers of the different groups at 35 and 49 d after immunization also revealed that the PPH-Nb-rAg@PM NP immunization group presented the highest serum titers of IgG (Fig. [148]5B). Fig. 5. [149]Fig. 5 [150]Open in a new tab PPH-Nb-rAg@PM NPs immunization elicits humoral immune response. (A) Schedule of mouse immunization and sampling. Mice were immunized with PBS, rAg, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs respectively twice with a 14-days intervals by the subcutaneous injection route. (B) Serum rAg-specific IgG titers at 28, 35, 49 d after immunization of each group was detected by ELISA. (C) Serum rAg-specific IgG2b, IgG2c, IgG3, IgA and IgM titers at 28 d after immunization of each group was determined by ELISA. (D and E) Serum rAg-specific IgG2a and IgG1 titers at 28, 35, 49 d after immunization of each group was detected by ELISA. (F) The IgG1/IgG2a ratio at 28, 35, 49 d after immunization of each group was calculated. (G) Standard virus-neutralizing assay. (H) IFA analysis of the effect of different mice immunized serum on ASFV p30 protein expression. (I) Fluorescence density analysis of (H) To assess the polarization of the immune response comprehensively, mouse serum IgG2b, IgG2c, IgG3, IgA and IgM titers against rAg at 28 d after immunization were determined via ELISA. Compared with those in the other groups, the titers of IgG2b, IgG2c, IgG3, IgA, and IgM in the groups immunized with the PPH-rAg-Nb@PM NPs were greater (Fig. [151]5C). In addition, compared with the other groups, the PPH-rAg-Nb@PM group presented higher IgG1 and IgG2a titers (Fig. [152]5D and E). Th1 cells and Th2 cells are two different subtypes of T cells, both of which play important roles in host defence and immune regulation and are associated with the production of different types of antibodies. Th1 cells are involved mainly in cell-mediated immune responses, such as the activation of macrophages and cytotoxic T cells. Th2 cells are involved mainly in humoral immunity, assisting in the activation of B cells and the production of antibodies. In mice, Th1 cells mainly produce IgG2a-type antibodies, whereas Th2 cells mainly produce IgG1-type antibodies. Subtype analysis by ELISA revealed that immunization with rAg, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM resulted in higher IgG1 than IgG2a levels (Fig. [153]5D and E), and the ratio of IgG1 to IgG2a was greater than 1.0, the PPH-rAg-Nb@PM NP immunization group presented the greatest IgG1-to-IgG2a ratio (Fig. [154]5F), implying a Th2-biased immune response. PPH-Nb-rAg@PM NPs induces ASFV-neutralizing antibodies After incubation of the NPs-immunized serially diluted mice serum (1:2, 1:4, 1:8, 1:16, 1:32) with 100 TCID[50] of ASFV suspension for 2 h at 37^oC, then the titer of each viral sample was assayed using TCID[50]. The results showed that for the rAg- or Nb-rAg-immunized group, when the dilution ratio of mice serum was 1:2, it could partially neutralize the activity of ASFV (Fig. [155]5G), the ratio was 1:4 for the PPH-Nb-rAg-immunized group, and 1:8 for the PPH-Nb-rAg@PM group (Fig. [156]5G). By detecting the expression of the ASFV p30 protein, it was observed that for the rAg-immunized group, a 1:2 dilution of mice serum partially suppressed ASFV p30 protein expression, and the Nb-rAg-immunized group showed the same results (Fig. [157]5H and I). For the PPH-Nb-rAg-immunized group, 1:4 but not 1:8 dilution of mice serum partially suppressed ASFV p30 protein expression, while the effective dilution increased to 1:8 in the PPH-Nb-rAg@PM-immunized group (Fig. [158]5H and I). These results indicate that PPH-Nb-rAg@PM induces higher neutralization antibodies against ASFV than other groups. PPH-Nb-rAg@PM NP immunization induced systemic cellular immune responses CD4 + and CD8 + T cells are key immune cells involved in the ASFV-specific adaptive immune response. CD4 + T cells play a critical role in promoting B-cell differentiation and sustaining the response of CD8 + cytotoxic T cells. Therefore, the T-cell responses in the spleens of immunized mice elicited by rAg, Nb-rAg, PPH-Nb-rAg, and PPH-Nb-rAg@PM NPs were further evaluated. At 28 d post vaccination, the proportion of CD3 + CD4 + T lymphocyte differentiation induced by Nb-rAg was 1.05 times greater than that induced by rAg, which induced by PPH-Nb-rAg was 1.12 times greater than that induced by Nb-rAg (Fig. [159]6A and B), and that induced by PPH-Nb-rAg@PM NPs was 1.52 times greater than that induced by PPH-Nb-rAg (Fig. [160]6A and B), which was the highest among all the groups. The proportion of CD3 + CD8 + T cells in the Nb-rAg group was 1.08 times greater than that in the rAg group, that in the PPH-Nb-rAg group was 1.11 times greater than that in the Nb-rAg group (Fig. [161]6C and D), and that in the PPH-Nb-rAg@PM NP group was the highest among the different groups, reaching 1.94 times greater than that in the PPH-Nb-rAg group (Fig. [162]6C and D). These results suggest that the PPH-Nb-rAg@PM NPs are able to elicit more effective CD4 + and CD8 + T-cell immune responses. Fig. 6. [163]Fig. 6 [164]Open in a new tab PPH-Nb-rAg@PM NPs immunization elicits cell immune response in vivo. (A and B) Representative flow cytometry plots and quantification analysis of the percentage of CD3 + CD4 + T cells of each group at 28 d post immunization with PBS, rAg, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs (n = 3). (C and D) Representative flow cytometry plots and quantification analysis of the percentage of CD3 + CD8 + T cells of each group at 28 d post immunization. (E) Shematic diagram of immunized mice splenocytes that were re-stimulated with rAg. (F and G) Analysis of IFN-γ secreting splenocytes clones re-stimulated with ASFV rAg protein were determined by ELISpot assay. (H and I) Analysis of IL-4 secreting splenocytes clones re-stimulated with ASFV rAg protein were determined by ELISpot assay. (J and K) Splenocytes isolated from each immunized group were re-stimulated with rAg for 60 h, and cell culture supernatants IL-4 and IFN-γ concentration were detected by ELISA, respectively. (L) The proliferation ability of lymphocyte of each group was detected using CCK-8 assay, and then the proliferation index was calculated based on the OD450 nm value Cellular immune responses benefit the generation of effective immunity and the clearance of invasive pathogens [[165]43]. Splenocytes isolated from immunized mice were restimulated with the rAg protein, followed by ELISpot measurement of IFN-γ- and IL-4-positive cells (Fig. [166]6E). Although the proportions of IFN-γ- and IL-4-secreting cells increased to some extent in each immunized group compared with those in the PBS control group at 28 d after immunization, the PPH-Nb-rAg@PM NP group presented the greatest number of IFN-γ- and IL-4-secreting cells (Fig. [167]6F-I). In addition, the IFN-γ and IL-4 levels in the splenocyte culture supernatants of each group were also detected via ELISA. Compared with the other groups, the PPH-rAg-Nb@PM NP group secreted the highest levels of extracellular IFN-γ (Fig. [168]6J) and IL-4 (Fig. [169]6K). The proliferation of lymphocytes reflects the host immune status [[170]44]. Thus, splenocytes isolated from each immunized group were restimulated with rAg for 72 h. CCK-8 assay results revealed that the proliferative capacity of lymphocytes isolated from PPH-Nb-rAg@PM NP-immunized mice was greater than that of the other groups (Fig. [171]6L). DC-targeting PPH-Nb-rAg@PM NPs promote BMDC maturation in vivo pXCR1-specific Nb was fused at the N-terminus of the ASFV rAg fragment, resulting in Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs capable of DC targeting. To further evaluate the effects of DC-targeting NPs on BMDC maturation in vivo, splenic DCs were isolated from immunized mice at 36 h after immunization, and an FCM assay was subsequently conducted to detect the phenotypes associated with DC maturation. The percentages of CD11c + CD80 + DCs in the PPH-rAg-Nb@PM NP-immunized group were 1.62 times greater than those in the PBS-immunized group, 1.41 times greater than those in the rAg-immunized group, 1.35 times greater than those in the Nb-rAg-immunized group, and 1.12 times greater than those in the PPH-Nb-rAg-immunized group (Fig. [172]7A). Analysis of CD11c + CD86 + and CD11c + MHCII + BMDCs also revealed that ASFV rAg conjugated with a specific Nb indeed promoted BMDC maturation more efficiently, especially the PPH-rAg-Nb@PM NPs, which exhibited the strongest ability to promote BMDC maturation (Fig. [173]7B and C). Fig. 7. [174]Fig. 7 [175]Open in a new tab PPH-Nb-rAg@PM NPs immunization promotes BMDCs maturation. The splenocytes of the PBS, rAg, Nb-rAg, PPH-Nb-rAg and PPH-Nb-rAg@PM NPs immunized mice were isolated at 36 h post immunization and then were subjected to FCM assay. Representative flow cytometry plots and quantification analysis of CD11c + CD80+ (A), CD11c + CD86+ (B), CD11c + MHCII + phenotype (C) BMDCs proportion were presented. (D-G) The isolated splenocytes were cultured for 48 h and supernatants were collected for detection of the secretion of IFN-γ (D), IL-12p70 (E), IL-4 (F), and IL-13 (G) using ELISA. (H and I) The isolated splenocytes were subjected to qPCR analysis of the mRNA expression of IRF4 and IRF8 that associated with BMDCs maturation Mature BMDCs can secrete various cytokines, which play important roles in the immune response, inflammatory response, and antigen presentation. For example, IFN-γ and IL-12 are capable of promoting Th1-type immune responses. IL-4 and IL-13 participate in the Th2-type humoral immune response. At 28 d postimmunization, the expression of these cytokines in isolated splenocytes after restimulation with rAg was further analyzed via ELISA. The results revealed that PPH-Nb-rAg@PM-immunized mice produced the highest levels of the Th1-type cytokines IFN-γ and IL-12p70 (Fig. [176]7D and E), as well as the Th2-type cytokines IL-4 and IL-13 (Fig. [177]7F and G). To further confirm that the PPH-Nb-rAg@PM NPs had greater immune efficiency in inducing BMDC maturation, the expression of several key regulators that participate in regulating BMDC maturation was assayed. Owing to the lack of available commercial antibodies, the expression levels of the IR4 and IR8 mRNAs were detected. qPCR results revealed that immunization with rAg, Nb-rAg or PPH-Nb-rAg indeed upregulated IR4 and IRF8 mRNA expression, especially in PPH-Nb-rAg@PM NP-immunized mice (Fig. [178]7H and I). These results suggest that, compared with ordinary ASFV rAg, PPH-Nb-rAg@PM NPs are able to promote splenic DC maturation more efficiently. PPH-Nb-rAg@PM NPs stimulate the activation of Tfhs and induce germinal center (GC) B cells and plasma cells DC maturation is able to induce a humoral immune response and facilitate the differentiation of T cells into Tfh cells. CD4 + Tfh cells reside in the spleen and are critical for the differentiation of GC-B cells into memory B cells and long-lived plasma cells [[179]45]. Therefore, CD4 + Tfh cells in the mouse spleen at 28 d postimmunization were investigated. Compared with the PBS treatment group, immunization with rAg increased the percentage of CD4 + CXCR5 + PD-1 + Tfh cells by 1.48-fold (9.72% vs. 6.55%), whereas the percentage of CD4 + CXCR5 + PD-1 + PD-1 + Tfh cells in the Nb-rAg-immunized group was 1.67-fold greater than that in the rAg group (16.24% vs. 9.72%), the percentage of CD4 + CXCR5 + PD-1 + Tfh cells in the PPH-Nb-rAg group was 1.21-fold greater than that in the Nb-rAg group (19.65% vs. 16.24%), the PPH-Nb-rAg@PM NPs group showed the highest percentage of CD4 + CXCR5 + PD-1 + Tfh cells and was 1.26-fold greater than that in the PPH-Nb-rAg group (24.71% vs. 19.65%) (Fig. [180]8A and B). Fig. 8. [181]Fig. 8 [182]Open in a new tab Detection of Tfh cells, GC-B cells and plasma cells from the immunized mice. (A) Gating strategy and representative flow cytometry plots of CD4 + CXCR5 + PD1 + Tfh cells, and quantitative analysis (B) of Tfh cells in the spleen (n = 3). (C) Gating strategy and representative flow cytometry plots of CD19 + CD95 + GL7 + GC-B cells in the spleen. (D) The percentage of GC-B cells (n = 3). (E) Gating strategy and representative flow cytometry plots of B220-CD138 + plasma cells in the spleen. (F) The percentage of plasma cells in the spleen (n = 3) The transcription regulatory factors Blimp-1, PD-1, and Bcl-6 are involved in regulating the production, development and differentiation of Tfh cells. Thus, the mRNA and protein levels of these transcription factors were further detected in the splenocytes of each immunized group. qPCR revealed that the PPH-Nb-rAg@PM NPs induced the highest levels of Blimp-1, PD-1, and Bcl-6 mRNA expression (Additional file 7: Figure [183]S7A-C). The western blotting results revealed that immunization with rAg, Nb-rAg or PPH-Nb-rAg promoted the expression of the Blimp-1, PD-1, and Bcl-6 proteins, and the PPH-Nb-rAg@PM NPs induced the highest expression levels of these proteins (Additional file 7: Figure [184]S7D). After antigen stimulation, GCs first form in secondary lymphoid organs such as the spleen, and then, antigen-specific B cells migrate to GCs to form GC-B cells [[185]46, [186]47]. With antigen stimulation and Tfh assistance, some GC-B cells differentiate into plasma cells or memory B cells [[187]48]. Thus, whether PPH-Nb-rAg@PM NP immunization could promote the production of GC-B cells was first determined at 28 d postimmunization. FCM revealed that the percentages of CD19 + CD95 + GL7 + phenotype GC-B cells were 0.30%, 0.44%, 0.65%, 2.3%, and 5.08% in the PBS-, rAg-, Nb-rAg-, PPH-Nb-rAg-, and PPH-Nb-rAg@PM NP-immunized groups, respectively. The PPH-Nb-rAg@PM group had the highest value, which was 11.55 times greater than that of the rAg group (Fig. [188]8C and D), indicating that PPH-Nb-rAg@PM NP immunization promoted the precursor production of plasma cells. Next, the proportion of B220-CD138 + phenotype plasma cells in the splenocyte population was determined. Compared with those of the PBS group (the proportion of plasma cells was 1.33%), the percentage of plasma cells with the B220-CD138 + phenotype of the mice immunized with rAg was 3.06%, which was 2.30 times greater than that of the PBS group; the percentages of the Nb-rAg, PPH-Nb-rAg groups were 6.94%, 10.39%, and were 2.27, 3.40 times greater than those of the rAg group; and the percentage of the PPH-Nb-rAg@PM NP-immunized group was the highest (13.41%), which was 4.38 times greater than that of the rAg group (Fig. [189]8E and F). PPH-Nb-rAg@PM NPs promoted memory B- and T-cell responses Memory B cells form in the GC after primary stimulation by antigens, and when re-exposed to the same specific antigen, they play an important role in producing faster and stronger antibody-mediated immune responses. Thus, splenic memory B cells were also analyzed (Fig. [190]9A). The results revealed that after rAg restimulation of splenocytes that were isolated at 28 d after immunization, the B220 + CD38 + IgG + IgD-GL7- phenotype memory B-cell proportion was markedly greater in the PPH-Nb-rAg@PM group than in the PBS, rAg, Nb-rAg and PPH-Nb-rAg groups (66.42% vs. 8.54%, 26.19%, 42.51%, and 54.91%, respectively) and was 2.54 times greater than that in the rAg group (Fig. [191]9B and C). Fig. 9. [192]Fig. 9 [193]Open in a new tab Detection of memory B cell, CD80 + memory B cells and central memory T cells in mice spleen. (A) Experimental schematic diagram. (B) Gating strategy and representative flow cytometry plots of B220 + CD38 + IgG + IgD-GL7- memory B cells. (C) Quantitative analysis of the percentage of memory B cells (n = 3). (D) Gating strategy and representative flow cytometry plots of CD80 + CD19 + IgD-IgG + memory B cells. (E) Quantitative analysis of the percentage of memory B cells (n = 3). (F) Gating strategy and representative flow cytometry plots of CD4 + CD44 + CD62L + central memory T cells. (G) The percentage of central memory T cells in immunized mice spleen CD80 is one of the key proteins expressed on the surface of memory B cells and is involved in regulating B-T-cell interactions as well as Tfh generation [[194]49, [195]50]. Thus, the expression of CD80 on the surface of the IgG + memory B cells was detected, and the results revealed that the percentage of CD19 + IgG + IgD-CD80 + memory B cells in the PPH-Nb-rAg@PM-immunized group was markedly greater than that in the PBS, rAg, Nb-rAg and PPH-Nb-rAg groups (71.51% vs. 14.13%, 28.18%, 31.21%, 37.15%, respectively) (Fig. [196]9D and E). Central memory T (T[CM]) cells circulate within the lymphatic system and proliferate and differentiate into effector T cells quickly to exert immune protection once they encounter the same antigen again. Boosting local and systemic memory T-cell responses is an effective strategy for obtaining long-term immunity, which is one of the pivotal features of an ideal vaccine. CD4 + CD44 + CD62L + T cells in the mouse spleen are considered typical TCMs with strong viability and can exist in the body for a long period. Thus, the splenocytes of each immunized group were isolated and restimulated with rAg, followed by FCM of the T[CM] proportion. After the boost immunization, although the proportions of CD4 + CD44 + CD62L + T cells were greater in the rAg, Nb-rAg and PPH-Nb-rAg groups than in the PBS treatment group (12.80%, 25.24%, 27.43% vs. 7.70%, respectively), the PPH-Nb-rAg@PM group presented the highest number of CD4 + CD44 + CD62L + T cells (32.23%), which was 2.52 times greater than that of the rAg-immunized group (Fig. [197]9F and G). The frequency of effector memory T (T[EM]) cells in mouse splenocytes was further investigated. The percentage of CD4 + CD44 + CD62L- phenotype T[EM] cells in the PPH-Nb-rAg@PM NP-immunized group was markedly greater than that in the PBS, rAg, Nb-rAg and PPH-Nb-rAg groups (21.33% vs. 3.39%, 6.36%, 9.36%, and 14.93%, respectively) (Additional file 8: Figure [198]S8). Single-cell transcriptomics analysis of immune cells in the spleen before and after immunization To further investigate the mechanism of the immune response induced by the PPH-Nb-rAg@PM NPs, scRNA-seq was performed. After initial quality control, 9959 and 8059 cells were obtained for single-cell transcriptome analysis in the PBS and PPH-Nb-rAg@PM groups, respectively. Uniform manifold approximation and projection (UMAP) identified eight cell subsets in the mouse spleen: T_NK cells, B cells, erythroblast cells, DCs, macrophages, neutrophils, endothelial cells, and plasma cells (Fig. [199]10A). Marker genes, including those in nonimmune and immune cell fractions, were analyzed to identify cell types. Among the identified cells, nonimmune cells primarily consisted of erythroblasts (marked by Spc24) and endothelial cells (marked by Cdh5). The immune cells included T_NK cells (marked by Cd3d), B cells (marked by Cd19), DCs (marked by Cd209a), macrophages (marked by Cd163), and plasma cells (marked by Jchain) (Fig. [200]10B). Analysis of the selected canonical markers for different cell subsets revealed that Cd3g, Cd3d and Nkg7, but not Ncam1, were highly expressed in T_NK cells; Cd19, Cd79a and Cd79b were highly expressed in B cells; Cdca5, Spc24 and Stmn1 were highly expressed in erythroblasts; Flt3 and Cd209a were highly expressed in DCs, while Itgam were highly expressed in neutrophils cells; Cd163, C1qa, C1qb and Csf1r were highly expressed in macrophages; Ly6g, Ncf1, Cd177, Sorl1, Csf3r and Retnlg were highly expressed in neutrophils; Pecam1 and Cdh5 were highly expressed in endothelial cells; and Mzb1 and Jchain were highly expressed in plasma cells (Fig. [201]10C), further confirming that the identified cell subsets were correct. Fig. 10. [202]Fig. 10 [203]Open in a new tab Single-cell analysis of splenocytes from mice immunized with PPH-Nb-rAg@PM. (A) UMAP analysis of splenocyte subsets. (B) Expression of cell-type-specific markers. (C) Markers for different cell subsets in mice splenocytes. (D) UMAP plot showed the expression profiles of mice spleens in PBS and PPH-Nb-rAg@PM groups, respectively. (E) Number of T_NK cells, B cells, erythroblast cells, DCs, macrophages, neutrophils, endothelial cells, plasma cells subsets of PBS and PPH-Nb-rAg@PM immunized mice spleens. The cell number of each population was calculated by multiplying the total number of isolated immune cells by the percentage of the population UMAP analysis of the quantitative changes in different cell subsets revealed that, compared with those in the PBS group, the numbers of T_NK cells, B cells, macrophages, and plasma cells in the spleens of the PPH-Nb-rAg@PM-immunized mice markedly increased (Fig. [204]10D and E), indicating that immunization with the PPH-Nb-rAg@PM NPs elicited extensive humoral and cellular immune responses in the mouse model. Compared with that in the PBS group, the population of neutrophils obviously decreased in the PPH-Nb-rAg@PM-immunized group (Fig. [205]10D and E), as neutrophils are associated with the inflammatory response. These results suggest that immunization with PPH-Nb-rAg@PM effectively inhibits the inflammatory response in vivo. PPH-Nb-rAg@PM vaccine induces protective immunity by enhancing immune cell activation On the basis of single-cell sequencing data, B-cell subtypes and T-cell subtypes were further analyzed. For the B-cell subtype, memory B cells and plasma cells were identified (Fig. [206]11A). Analysis of marker molecules expressed on the surface of B-cell subtypes further confirmed that memory B cells (marker protein Ms4a1, Fig. [207]11B) and plasma cells (marker protein Jchain) were identified as expected (Fig. [208]11C). In addition, the expression patterns of surface proteins also revealed that Aim2 and Ms4a1 were expressed mainly on memory B cells, whereas Jchain and Mzb1 were expressed mainly on plasma cells (Fig. [209]11D). Moreover, in the spleen at 28 d postimmunization, significant increases in the numbers of memory B cells and plasma cells were observed (Fig. [210]11E and F). These results indicate that the PPH-Nb-rAg@PM nanovaccine enhances the magnitude of humoral immune responses, which is in accordance with the results shown in Figs. [211]8E and [212]9B. Fig. 11. [213]Fig. 11 [214]Open in a new tab Single-cell analysis of B and T cell subtypes from mice immunized with PPH-Nb-rAg@PM. (A) UMAP analysis of B cells subsets. (B) Expression of memory B cell-specific marker. (C) Expression of plasma cell-specific marker. (D) Markers for memory B cells and plasma cells in mice splenocytes. (E) UMAP plot showed the expression profiles of memory B cells and plasma cells in PBS and PPH-Nb-rAg@PM immunized mice, respectively. (F) Number of memory B cells and plasma cells subsets of PBS and PPH-Nb-rAg@PM immunized mice spleens. (G) UMAP analysis of T cells subsets. (H-K) Expression of CD4 + T cells (H), CD8 + T cells (I), NK cells (J), and NKT cells (K) cell-specific marker. (L) Markers for CD4 + T cells, CD8 + T cells, NK cells, and NKT cells in mice splenocytes. (M) UMAP plot showed the expression profiles of CD4 + T cells, CD8 + T cells, NK cells, and NKT cells in PBS and PPH-Nb-rAg@PM immunized mice, respectively. (N) Number of CD4 + T cells, CD8 + T cells, NK cells, and NKT cells subsets of PBS and PPH-Nb-rAg@PM immunized mice spleens. (O) The cross-talk between B cells, T_NK cells, plasma cells, neutrophils, and erythroblast The T_NK cell subtypes were further analyzed via scRNA-seq. UMAP analysis revealed that the identified T_NK cell subtypes could be further divided into CD4 + T cells, CD8 + T cells, NK cells and NKT cells (Fig. [215]11G). UMAP analysis of the expression patterns of surface marker proteins confirmed that CD4 + T cells (marker protein Cd4, Fig. [216]11H), CD8 + T cells (marker protein Cd8a, Fig. [217]11I), NK cells (marker protein Nkg7, Fig. [218]11J) and NKT cells (marker protein Gzmb, Fig. [219]11K) were correctly identified. Analysis of the selected canonical markers for different cell subsets revealed that Cd3d, Cd3e, Cd3g and Cd4 were highly expressed in CD4 + T cells; Cd3d, Cd3e, Cd3g and Cd8a were highly expressed in CD8 + T cells; Nkg7 and Gzmb were highly expressed in NK cells; and Cd3d, Cd3e, Cd3g and Nkg7 were highly expressed in NKT cells (Fig. [220]11L), confirming that the T-cell subtypes were correctly identified. UMAP analysis revealed that at 28 d postimmunization, the numbers of CD4 + T cells, CD8 + T cells and NK cells in the PPH-Nb-rAg@PM immunization group were markedly greater than those in the PBS control group, which was consistent with the FCM results shown in Figs. [221]4B and [222]8A and C. However, the number of NKT cells in the PPH-Nb-rAg@PM group was lower than that in the PBS group (Fig. [223]11M and N). Cell-cell communication network analysis also suggested that mouse spleen B cells interacted more tightly with DC clusters, T_NK cell clusters, and plasma cell clusters after PPH-Nb-rAg@PM immunization (Fig. [224]11O). Taken together, these results suggest that the PPH-Nb-rAg@PM nanovaccine effectively enhances the B and T-cell immune responses in vivo. Discussion There is no vaccine or drug available for ASF globally, and the prevention and control of the disease relies on strict biosecurity measures, early diagnosis and culling of infected pigs. The subunit vaccine is a relatively safe vaccine composed of a variety of viral proteins or peptides that are immunogenic and can induce neutralizing antibodies in the body. However, immune-related viral proteins have not been fully characterized and are composed of only a limited number of viral proteins, such as p30, p72, p54 and CD2v, which are not enough to induce the body to produce strong neutralizing antibodies. The immunoprotective effect was low. DC-targeted vaccines have become a research focus in the field of vaccine development for animal epidemic diseases, although much of this research is still in its infancy. Specific nanobody-mediated ASFV antigen targeting to DCs is a promising strategy for the development of DC-targeted ASFV vaccines. The present study developed a biomimetic nanovaccine platform based on PM-decorated phosphorus dendrimer/Nb-rAg NPs for the development of a candidate vaccine for ASFV. By coupling specific Nbs with ASFV recombinant antigen epitopes, the antigen recognition ability of DCs was significantly enhanced. Compared with mice immunized with ordinary ASFV antigens, those immunized with Nb-rAg exhibited greater specific humoral or cellular immune responses. Among the many different delivery systems, the PPH, which is characterized by a highly branched three-dimensional structure, a functionalize surface and a uniform molecular weight, has received much attention [[225]51]. PPH can induce NK cell proliferation by specifically inhibiting CD4 + T lymphocyte proliferation, and NK cells play an important role in antimicrobial infection [[226]31]. Compared with the Nb-rAg group, the combination of the Nb-rAg complex with the PPH significantly promoted the proliferation of NK cells in immunized mice; compared with the Nb-rAg group, the PPH-Nb-rAg group presented a greater number of NK cell populations, and the PPH-Nb-rAg group also significantly promoted the proliferation of NK cells in vitro, which may promote the anti-ASFV infection ability of immunized pigs. In addition, the decoration of preactivated macrophage membranes prevents the NPs from being engulfed by macrophages, allowing them to have a longer cycle time in the body and inducing a stronger immune response. As demonstrated by the larger population numbers of GC B cells, memory B cells and plasma cells, higher levels of specific antibodies were induced by the PPH-Nb-rAg@PM NPs. In addition to their role in humoral immune responses, T cells play important roles in various viral infections. CD4 + T cells mediate the production of antibodies by B cells, coordinate the response of other types of immune cells, and directly initiate an immune response to infectious agents [[227]52]. CD4 + T cells differentiate into Th2 and Th1 cells that drive the adaptive immune response, and CD8 + T cells target infected cells and clear infection sites, mainly through perforin, granzyme, and the FasL pathway [[228]53]. The present results revealed that immunization with PPH-Nb-rAg@PM resulted in the highest levels of CD4 + and CD8 + T cells, and ELISpot resulted in the highest secretory levels of IFN-γ and IL-4 in mouse splenocytes, confirming that PPH-Nb-rAg@PM is superior to other types of vaccines in inducing a cellular immune response, as the production of IFN-γ and IL-4 by antigen-stimulated PBMCs is viewed as an indicator of vaccine immunogenicity. Single-cell sequencing can be used to analyze the immune cell types of mice directly. According to the single-cell sequencing results, after immunization with PPH-Nb-rAg@PM, the proportions of immune cells, such as T_NK cells, B_cells, DC macrophages and plasma_cells, significantly increased compared with those in the PBS control group, implying that the PPH-Nb-rAg@PM nanovaccine had a good immune effect. Further analysis of the B-cell subtype or T-cell subtype also suggested that immunization with PPH-Nb-rAg@PM elicited strong immune B and T-cell responses, which further confirmed the potential of PPH-Nb-rAg@PM as a candidate vaccine for ASFV. Owing to the high cost and biosafety factors, the current study did not investigate the immunoprotective potential of PPH-Nb-rAg@PM nanovaccines in ASFV-infected pigs, which is also a shortcoming. Besides, the role and mechanism of TLRs expressed on the surface of macrophage membrane in PPH-Nb-rAg@PM-induced humoral and cellular immunose is still unknown, and this also needs further studies in the future. However, multiepitope vaccine subunit vaccines have the unique advantage of high safety, which is not possessed by other types of vaccines. The PPH developed in this study integrates several reported epitopes with viral neutralizing activity. Both in vivo and in vitro experiments have shown that nanovaccines can induce higher levels of humoral and cellular immune responses than conventional vaccines can, and can produce higher levels of immunomodulators. It has great potential for development into a new ASFV vaccine and warrants further research. This study also provides a platform for the development of other viral subunit vaccines. Conclusions The present study developed a nanovaccine, PPH-Nb-rAg@PM, which recombines multiple ASFV neutralization epitopes and then conjugates them with high-affinity, specific Nbs against the DC antigen receptor XCR1. The recombinant protein Nb-rAg, which targets DCs, was expressed and purified via a prokaryotic expression system, and then, Nb-rAg and PPH were coupled to obtain PPH-Nb-rAg. Then, the PPH-Nb-rAg NPs were coated with ASFV-preactivated macrophage membranes to construct the PPH-Nb-rAg@PM biomimetic nanovaccine. Then, the mice were immunized with the nanovaccine and PBS; rAg, Nb-rAg and PPH-Nb-rAg were used as control groups. Compared with the other control groups, the PPH-Nb-rAg@PM nanovaccine induced a stronger humoral immune response, cellular immune response, greater NK cell proliferation ability, and greater numbers of memory B cells, memory T cells, and plasma cells, indicating that the developed nanovaccine has potential as an effective candidate vaccine for ASFV prevention and control. Electronic supplementary material Below is the link to the electronic supplementary material. [229]Supplementary Material 1^ (4.7MB, docx) [230]Supplementary Material 2^ (28KB, docx) Acknowledgements