Abstract Background Flavivirus is a highly prevalent and outbreak-prone disease, affecting billions of individuals annually and posing substantial public health challenges. Vaccination is critical to reducing the global impact of flavivirus infections, making the development of a safe and effective vaccine a top priority. The self-assembled pan-epitope vaccine presents key advantages for improving immunogenicity and safety without relying on external vectors or adding immunomodulatory elements, both of which are essential for successful vaccine development. Results In this study, the pan-epitope peptide TBT was combined with adjuvant CpG to form the TBT-CpG nanovaccine (TBT-CpG NaVs), which was found to be spherical, uniform in shape, and demonstrated strong serum stability. In vitro studies showed that the TBT-CpG NaVs were efficiently taken up and internalized by bone marrow-derived dendritic cells (BMDCs). Flow cytometry and transcriptomic analysis indicated that the antigens were effectively presented to antigen-presenting cells (APCs) via the MHC II pathway, which facilitated BMDCs maturation and promoted the release of pro-inflammatory cytokines IL-1β, TNF-α, and IL-6. In vivo studies confirmed that TBT-CpG NaVs enhanced antigen-specific IgG levels, significantly increased IFN-γ and IL-4 expression in spleen cells, and offered protective effects against Dengue virus (DENV) and Zika virus (ZIKV) infections. Safety evaluations revealed no hepatotoxicity and no significant organ damage in immunized mice. Conclusion The self-assembled candidate nanovaccine TBT-CpG NaVs effectively activates BMDCs and triggers a targeted immune response, providing antiviral effects against DENV and ZIKV. This vaccine demonstrates good immunogenicity and safety, establishing a promising foundation and a new strategy for the development of safe and effective vaccines. Graphical Abstract [46]graphic file with name 12951_2024_3031_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-03031-0. Keywords: Flavivirus, Pan-epitope peptide, CpG, Nanovaccine Introduction Flaviviruses, a group of viruses transmitted by arthropods, include several significant human pathogens, such as Dengue virus (DENV), Zika virus (ZIKV), Yellow fever virus (YFV), and West Nile virus (WNV) [[47]1, [48]2]. These viruses threaten billions of people worldwide, representing an increasing global health risk [[49]3, [50]4]. DENV, with its four serotypes (DENV1-4), is the most widespread mosquito-borne virus, while ZIKV, known for its severe teratogenic and neurotoxic effects, garners particular attention due to their substantial impact on public health [[51]5]. Vaccination remains a critical strategy for controlling infectious diseases, with ample evidence of its effectiveness. For instance, the success of COVID-19 vaccines in reducing disease severity and incidence underscores the essential role of vaccines in mitigating severe outcomes and hospitalizations. Globally, vaccines continue to play an irreplaceable role in controlling communicable diseases. Currently, no vaccines for ZIKV have received approval, and only two live-attenuated dengue vaccines—Dengvaxia (CYD-TDV) and DENVax (TAK003)—are clinically approved [[52]6]. Although live-attenuated vaccines retain a nearly complete virus structure to trigger mucosal, humoral, and cellular immune responses [[53]7, [54]8], their protective efficacy against the four DENV serotypes remains inconsistent. Additionally, the use of live pathogens carries a risk of pathogenicity recovery [[55]9, [56]10]. In contrast, a new class of epitope-based subunit vaccines, which contain only specific pathogen antigen fragments, offers enhanced safety and scalability for large-scale production [[57]11]. Epitope-based peptide vaccines target specific regions of a pathogen’s proteins, known as epitopes, ensuring a focused immune response against critical areas of the pathogen, thereby improving efficacy and reducing the risk of off-target effects [[58]12, [59]13]. The design and synthesis of peptide vaccines can be rapidly accomplished once a pathogen’s genetic information is available, a crucial advantage during outbreaks of new infectious diseases. The primary limitation of epitope-based subunit vaccine is their low immunogenicity, due to the absence of a complete organism [[60]14]. This limitation can be addressed by combining subunit vaccines with adjuvants and enhancing the capacity of dendritic cells (DCs) to uptake and present antigen epitopes, both of which are effective for boosting antiviral immune responses and achieving long-lasting protective efficacy [[61]15–[62]17]. CpG, a widely used adjuvant, act as a TLR9 agonist that promotes antigen endocytosis to enhance the immune response, with a safety profile similar to conventional vaccines [[63]18, [64]19]. Among CpG options, ODN 1826 (5’-TCC ATG ACG TTC CTG ACG TT-3’) is a well-known immunological adjuvant, activating the innate immune response and improving the function of professional antigen-presenting cells (APCs) with proven safety [[65]20]. Furthermore, the programmability and chemical functionalization of DNA sequences allow for the formation of diverse nanostructures [[66]21, [67]22]. Co-delivering antigens with CpG ODN in nanocarriers promotes a robust immune response and enhances antitumor effects, both within and outside these carriers [[68]23]. As professional APCs, DCs play a central role in immune induction by capturing and processing antigens. Immature DCs, in particular, exhibit high migratory abilities and preferentially uptake virus-sized particles (20–200 nm) [[69]24–[70]26]. Linking target peptide epitopes to nanoparticles aids in the maturation of bone marrow-derived dendritic cells (BMDCs) and facilitates the organized presentation of multivalent epitopes [[71]27]. This approach enhances antigen bioavailability and effectively activates both innate and adaptive immune responses, which is critical for developing an effective candidate vaccine [[72]28, [73]29]. Self-assembled nanovaccines, which combine antigens with adjuvants, have proven to be an excellent vaccine platform, offering several advantages: (1) They avoid the inclusion of unnecessary components, such as delivery carriers, thereby reducing the risk of carrier-related toxicity thanks to their simple structure [[74]30]; (2) They ensure the co-delivery of antigens and adjuvants directly to APCs, inducing targeted immune responses and minimizing the risk of immune tolerance [[75]21, [76]31, [77]32]; and (3) They enhance antigen stability, prolong antigen release, and support sustained immune responses over time [[78]28]. Various types of nanovaccine have shown promise in antiviral and antitumor studies, with self-assembled peptide nanoparticles due to their high antigen load and lack of external carriers [[79]33–[80]35]. However, no research has yet explored a self-assembled co-delivery system specifically based on flavivirus epitope antigens and adjuvants. In this study, we developed a self-assembled nanovaccine to combat flavivirus, using a conjugate of the pan-epitope peptide TBT with the adjuvant CpG. The hydrophobic TBT was conjugated with the hydrophilic CpG using DTDP and the reductant TCEP, resulting in self-assembly through hydrophobic and electrostatic interactions to form the TBT-CpG nanovaccines (TBT-CpG NaVs). This nanovaccine co-delivers antigens and adjuvants to dendritic cells (DCs), promoting the maturation of bone marrow-derived dendritic cells (BMDCs) and stimulating cytokine secretion. TBT-CpG NaVs effectively activated both cellular and humoral immunity against the flavivirus with excellent biosafety, establishing a promising foundation for the next generation of flavivirus vaccine candidates. Materials and methods Preparation of TBT-CpG NaVs The synthesis of self-assembled nanovaccine was carried out as follows: CpG ODN (Sangon Biotech, China) was mixed with Tris (2-carboxyethyl) phosphine (TCEP) solution at a molar ratio of 1:100 for 2 h at 25 °C. This mixture was then combined with 4,4’-Dithiodipyridine (DTDP) solution at a molar ratio of 1:1000 for 30 min at 25 °C. The resulting product was analyzed with mass spectrometer (Thermo, USA) and purified using an NAP-5 nucleic acid purification column (Cytiva, USA) to remove excess TCEP and DTDP. The pan-epitope peptide TBT (KYVKQNTLKLAT-GG-VDRGWGNGCGLFGKG-LL-LEYIPEITLPVIAALSIAES, Chinese Peptide, China) was then mixed with purified products at molar ratios of 1:1, 2:1, 3:1, 4:1, 5:1 and 6:1 for 30 min at 25 °C to produce self-assembled nanovaccine TBT-CpG NaVs with varying loading degrees. Gel electrophoresis experiment The conjugated products of TBT-CpG and free CpG were loaded into a 1% agarose gel and electrophoresed for 80 min at 60 V. Free CpG, TBT, and TBT-CpG were also loaded into a 15% native polyacrylamide gel electrophoresis (PAGE) and run for 2 h at 80 V. Following electrophoresis, the gel was stained with a 3× Gel Red staining solution in the dark for 30 min at 25 °C. Visualization and analysis were conducted using a Fusion SoloS gel imager. Characterization of TBT-CpG NaVs To characterize the TBT-CpG NaVs, 20 µL of the conjugated products were diluted in PBS at a volume ratio of 1:50. The average particle size and Zeta potential of the sample were measured at 25 °C using a Zetasizer Nano-ZS90 (Malvern Instruments Ltd., UK). For morphological analysis, self-assembled TBT-CpG NaVs were prepared by negative staining transmission electron microscopy (TEM). Briefly, 10 µL of the sample was dropped onto a copper grid and absorbed for 10 min. After removing excess liquid, the sample was stained with 10 µL of uranyl acetate for 1 min. Once dried, the sample’s dispersion and morphology were observed with JEM 1200EX transmission electron microscope. In vitro pan-epitope peptide TBT Release Study The release profile of pan-epitope peptide TBT from TBT-CpG NaVs was evaluated through dialysis, with TBT labled by Cy5. Briefly, TBT-CpG NaVs were placed in dialysis bags (Viskase, USA), which were securely sealed. The dialysis bags were immersed in 50 mL of release medium, consisting of phosphate-buffered saline solution buffer with 1mM glutathione (GSH), and incubated at 37 °C with continuous shaking at 100 rpm. 0.5 mL of dialysate was collected and replaced with an equal volume of fresh release medium to maintain sink conditions. The fluorescence intensity of TBT was measured using a SYNERGY H1 fluorescence spectrophotometer (BioTek, USA), with a series of diluted Cy5-labeled TBT used to establish standard curves. Serum stability study of TBT-CpG NaVs To assess serum stability, TBT-CpG NaVs and a control group of free CpG were treated with DMEM medium containing 10% FBS for 24 h, 48 h, and 72 h. After each time point, samples were loaded into a 1% agarose gel and electrophoresed for 80 min at 60 V to evaluate their stability. Isolation and cytotoxicity analysis of mouse bone marrow-derived dendritic cells (BMDCs) BMDCs were isolated under sterile conditions from the femurs and tibias of mouse and cultured in RPMI1640 medium with 10% FBS, 1% penicillin/streptomycin, 20 ng/mL mGM-CSF and 10 ng/mL mIL-4 (Novoprotein, China) at 37 °C in a cell incubator. Half of the medium was replaced on the 2nd and 4th days, and immature cells were collected for cytotoxicity analysis on the 6th day. BMDCs were seeded in a 96-well plate at a density of 1 × 10^4 cells/well and incubated overnight. TBT, TBT + CpG, and TBT-CpG NaVs were then added at final concentrations of 50, 100, 200, 400, and 600 nM, with PBS as a control. Cell viability was determined using the Cell Counting Kit-8 (Beyotime, China) after 24 h at 37 °C. Absorbance at 450 nm was measured using a microplate reader. Cell viability (%) was calculated using the formula: (Atreat/Acontrol) × 100%, where Atreat represented the absorbance of TBT/TBT + CpG/TBT-CpG NaVs treated cells, and Acontrol represented the absorbance of the control group cells. In Vitro uptake and activation of BMDCs BMDCs were seeded at in a 24-well culture plate at a density of 1 × 10^5 cells/well, and incubated overnight. Cy5-labeled TBT, TBT + CpG, and TBT-CpG NaVs were co-incubated with BMDCs for 2–24 h. After 2 h of incubation, the samples were fixed with 4% paraformaldehyde for 15 min and stained with DAPI for 5 min. The fluorescence signal was visualized under a confocal microscopy (ZEISS, LSM880, Germany). The percentage of Cy5-positive cell was analyzed via flow cytometer (BD FACS Celesta, USA). After 24 h of incubation, the culture supernatant was collected to measure inflammatory cytokines (TNF-α, IL-1β, and IL-6) using ELISA kits (Thermo Fisher, USA). Cells were then stained with antibodies CD11c-APC, CD80-PE, CD86-FITC, and MHC-II-PE/Cyanine7 (BioLegend, USA). The expression of surface markers CD11C, co-stimulatory molecules CD80 and CD86, and MHC II on BMDCs was detected by flow cytometry, with BMDCs cultured alone serving as a negative control. RNA-Seq and data analysis Sample RNA was reverse transcribed using the Illumina NovaSeq Reagent Kit to construct a cDNA library, followed by high-throughput sequencing on the Illumina NovaSeq 6000 platform. Raw sequencing data were processed with fastp software to obtain high-quality clean reads for further analysis. These clean reads were aligned to the mouse genome (Mus_musculus. GRCm39) using Hisat2. Gene and transcript levels were quantified with RSEM software. Differential gene expression analysis was conducted using DESeq2, identifying genes as differentially expressed if they met both criteria (p < 0.05 & |log2FC| ≥ 1). GO enrichment analysis was performed using Goatools, and KEGG pathway enrichment analysis was conducted with KOBAS. Animal immunization Female Balb/c mice aged 6–8 weeks were purchased from the Army Medical University Laboratory Animal Center. All animal procedures were approved by the Army Medical University Animal Welfare and Ethics Committee (AMUWEC20240036). After a 7-day acclimation period, the mice were randomly assigned to four groups (PBS, TBT, TBT + CpG, and TBT-CpG NaVs), with six mice per group. Vaccination were administered via subcutaneous injection on days 0, 14, and 28, with each dose containing 30 µg TBT in a volume of 200 µL. Serum samples were collected on days 35 and 56 post-initial immunization for IgG antibody analysis. Seven days after the final immunization, organs were aseptically collected for Enzyme-linked Immunospot Assay (ELISPOT) and safety evaluation. IgG antibody titer detection TBT was dissolved in coating buffer at a final concentration of 5 µg/mL, with 100 µL of antigen solution per well in a 96-well plate, and incubated overnight at 4 °C. The plate was washed five times with wash buffer containing 0.1% Tween 20 to remove unbound antigen. Wells were blocked with 200 µL of blocking solution (carbonate buffer containing 5% bovine serum albumin) and incubated at 37 °C for 2 h to prevent non-specific binding. After washing five times, diluted serum samples were incubated at 37 °C for 2 h. Following additional washing, 100 µL of horseradish peroxidase-labeled anti-mouse IgG secondary antibody at a dilution of 1:5000 was added and incubated at 37 °C for 1 h. The reaction was developed with TMB substrate solution for 10 min, stopped with 100 µL of 2M H[2]SO[4], and absorbance was measured at 450 nm using a microplate reader Synergy H1. Enzyme-linked immunospot assay (ELISPOT) Seven days after the final immunization, mouse spleen was aseptically harvested to prepare single-cell suspension. The cell suspension was adjusted to a concentration of 5 × 10^6 cells/mL with RPMI1640 medium, and 100 µL per well was added to pre-coated 96-well plates. Peptide TBT was used as a stimulant, and cells were incubated at 37 °C for 36 h. Cytokines IFN-γ and IL-4 were detected using ELISPOT kits (Dakewei, China), and spots were analyzed with an immune spot microscope. In vivo viral challenge protection experiment On day 35 after the initial immunization, blood samples were collected from each group and centrifuged at 1000 g for 10 min to isolate serum. The serum was then inactivated at 56 °C for 30 min, diluted with PBS, and mixed with 10 µL of DENV-2, DENV-4, or ZIKV at 37 °C for 1 h. The virus-serum mixture was injected intracranially into suckling mice using a microsyringe, and survival was monitored for 15 days post-infection. Bio-safety analysis On day 35 post-immunization, mice were euthanized to collect blood and major organs (heart, liver, spleen, lungs, kidneys, brain. Serum levels of ALT and AST were measured using biochemical assay kits (Solarbio, China) following the manufacturer’s instructions. Organs were preserved in 4% paraformaldehyde and prepared for hematoxylin-eosin staining. Statistical analysis All data are presented as mean ± SD. Statistical analyses were performed using GraphPad Prism 9.0 software, employing Student’s t-test or one-way analysis of variance (ANOVA). A P-value < 0.05 is considered statistically significant (*P < 0.05, **P < 0.01, ***P < 0.001, **** P < 0.0001). Results Preparation and characterization of TBT-CpG nanovaccines (NaVs) The thiolated CpG ODN (CpG-SH) was coupled with DTDP to creat a new product CpG-DTDP, with its molecular weight displayed in Fig. [81]S1. The purified CpG-DTDP was then conjugated with the flavivirus pan-epitope peptide TBT via disulfide bonds, resulting in the TBT-CpG product. To quantify the binding capacity between the antigenic peptide TBT to the adjuvant CpG ODN, TBT was mixed with CpG ODN at different molar ratios, and unbound CpG ODN was monitored via agarose gel electrophoresis. Uptake of the peptide by DC2.4 cells was visualized using the fluorescence microscopy, revealing a marked increase in cellular uptake for TBT when coupled with CpG ODN compared to free TBT (Fig. [82]S2). Full binding of CpG ODN to TBT occurred at a molar ratio exceeding 3:1 (Fig. [83]1A). Fig. 1. [84]Fig. 1 [85]Open in a new tab Preparation and characterization of TBT-CpG NaVs. A Agarose gel electrophoresis showing the conjugation reaction between TBT and CpG ODN at varying molar ratios. Lane 1: Free CpG ODN; Lanes 2–7: Conjugation reactions with TBT and CpG ODN at different molar ratios. B and C Dynamic Light Scattering (DLS) analysis for particle size (B) and zeta potential (C) of TBT-CpG NaVs at different TBT-to-CpG ODN ratios. D Transmission Electron Microscopy (TEM) image displaying the morphology of TBT-CpG NaVs. E Native polyacrylamide gel electrophoresis (PAGE) with Gel Red staining, comparing TBT-CpG NaVs at a 4:1 molar ratio with a physical mixture of TBT and CpG ODN at the same ratio (TBT + CpG). F Agarose gel electrophoresis results for serum stability testing of TBT-CpG NaVs and the physically mixed TBT + CpG at 24, 48, and 72 h. Data are represented as mean ± SD (n = 3). Statistical analysis was performed using Student’s t-test (B, C). ns, p > 0.05; ***p < 0.001 To identify the optimal TBT to CpG ODN binding ratio, the conjugated products at various ratios were analyzed using the Dynamic Light Scattering (DLS) for particle size and zeta potential. TBT-CpG NaVs formed at a 4:1 molar ratio exhibited a smaller diameter of 153.8 ± 1.7 nm with a PDI of 0.287 ± 0.023 (Fig. [86]1B and Fig. [87]S3) and the highest zeta potential at 28.1 ± 1.7 mV (Fig. [88]1C), which is favorable for cellular uptake. Following optimizing the nanovaccine synthesis, the self-assembled nanovaccine TBT-CpG NaVs were further characterized. Transmission electron microscopy revealed that TBT-CpG NaVs were spherical and uniformly distributed (Fig. [89]1D). Compared with free CpG ODN, TBT-CpG NaVs showed a reduced migration rate with no detectable residual CpG, indicating the adjuvant CpG was bound successfully to TBT via disulfide bonds, resulting in a high loading rate (Fig. [90]1E). The cumulative release of TBT was 57.2 ± 1.7% over 48 h under 1mM GSH (Fig. [91]S4A). These results confirmed the successful development of a self-assembled spherical nanovaccine with high encapsulation efficiency and slow antigen peptide TBT released. For practical application, TBT-CpG NaVs, as a novel vaccine delivery system, should be physiological stability. Serum stability was assessed through agarose gel electrophoresis, the result revealed that the self-assembled TBT-CpG NaVs have significantly enhanced stability in DMEM medium with 10% FBS compared to the physically mixed TBT + CpG group (Fig. [92]1F). Stability was also maintained in DMEM medium containing 30% and 50% FBS (Fig. [93]S4B). TBT-CpG NaVs facilitate rapid uptake and internalization by BMDCs Flow cytometry analysis revealed that TBT-CpG NaVs exhibited a significantly higher antigen uptake capacity compared to the TBT and TBT + CpG groups (Fig. [94]2A). To further investigate the immunological mechanism of the TBT-CpG NaVs vaccine, Cy5-labeled TBT was employed to track the internalization of TBT-CpG NaVs (Fig. [95]2B). After co-culturing 200 nM Cy5-TBT with BMDCs for 2 h, TBT-CpG NaVs demonstrated markedly increased red fluorescence, primarily localized in the cytoplasm, compared to the free TBT and TBT + CpG groups. These findings suggested that TBT-CpG NaVs are rapidly taken up and internalized by BMDCs, potentially enhancing the effectiveness of immune response activation. Fig. 2. [96]Fig. 2 [97]Open in a new tab Uptake and internalization of TBT-CpG NaVs by BMDCs. A Flow cytometry anslysis to evaluate the uptake efficiency of different nanoparticles in BMDCs after 2 h at a concentration of 200 nM. B Confocal laser scanning microscopy images showing the internalization of different nanoparticles in BMDCs at a concentration of 200 nM after 2 h. TBT is labeled with Cy5 (red), and cell nuclei are stained with DAPI (blue). Scale bars represent 20 μm. Data are shown as mean ± SD (n = 3). Statistical analysis was performed using one-way ANOVA. ****p < 0.0001 TBT-CpG NaVs induce maturation of BMDCs and promote cytokine secretion To evaluate the effect of TBT-CpG NaVs on the activation and maturation of BMDCs, BMDCs from C57BL/6 mouse were cultured with mGM-CSF and mIL-4 for 6 days before being treated with 200 nM TBT-CpG NaVs for 24 h. Flow cytometry was employed to measure the expression levels of cell surface molecules MHC II and co-stimulatory molecules CD80 and CD86. CD11C, a marker commonly used for dendritic cell identification, was expressed over 95% of cells in all groups, confirming the successful isolation of high purity BMDCs (Fig. [98]S5A). The percentage of CD80^+CD86^+BMDCs was significantly higher in the TBT-CpG NaVs group than in the TBT + CpG group (Fig. [99]3A and Fig. [100]S5B). Additionally, TBT-CpG NaVs treatment led to increased MHC II expression following antigen stimulation (Fig. [101]3B and Fig. [102]S5C). ELISA analysis of the cell culture supernatant revealed that TBT-CpG NaVs significantly upregulated the levels of pro-inflammatory factors IL-1β, TNF-α, and IL-6 (Fig. [103]3C-E). These results demonstrated that TBT-CpG NaVs enhanced antigen presentation through the MHC II pathway, thereby stimulating the maturation of dendritic cells. Fig. 3. [104]Fig. 3 [105]Open in a new tab TBT-CpG NaVs induce maturation of BMDCs and promote cytokine secretion. Flow cytometry was used to measure the expression of CD80^+CD86^+ (A) and MHC II^+ (B) in BMDCs, after co-culturing with different stimuli for 24 h. The bar graph of (A) represented the statistical analysis of CD80^+CD86^+. ELISA was used to assess cytokine levels in the supernatant of cells stimulated by different treatments: IL-1β (C), TNF-α (D), and IL-6 (E). Data are presented as mean ± SD (n = 3). Statistical analysis was conducted using one-way ANOVA. ns, p > 0.05; *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001 Transcriptomic analysis of BMDCs stimulated by TBT-CpG NaVs To explore the underlying mechanisms by which TBT-CpG NaVs promote BMDC maturation, transcriptomic analysis was performed on BMDCs treated with either TBT alone or TBT-CpG NaVs. Differentially expressed gene (DEG) analysis revealed substantial gene expression changes with TBT-CpG NaVs compared to free TBT, with 394 genes upregulated and 58 genes downregulated, showing a predominance of upregulated genes (Fig. [106]4A). Fig. 4. [107]Fig. 4 [108]Open in a new tab Transcriptome analysis of BMDCs stimulated by TBT-CpG NaVs. A Volcano plot showing differentially expressed genes, with upregulated genes marked in red and downregulated genes in blue. B GO enrichment analysis of upregulated genes, where the vertical axis represented the GO term and the horizontal axis represented the Rich factor. Dot size indicates the number of genes in each GO term, and dot color corresponded to different range of adjust p-values. C KEGG enrichment analysis of the upregulated genes. The right side shows pathways significantly enriched in differential genes, while the left side lists genes in each pathway, ordered by log2 Fold Change (log2FC). A higher log2FC indicates a greater fold increase in gene expression. D Reactome enrichment analysis of upregulated genes. The vertical axis represents Reactome pathways. The upper horizontal axis shows the number of genes mapped to each pathway, while the lower axis represents the significance level of enrichment Gene Ontology (GO) enrichment analysis indicated that these differential genes were primarily involved in myeloid dendritic cell activation, antigen processing and presentation of exogenous peptide antigen via MHC class II, and the regulation of T cell proliferation and differentiation (Fig. [109]4B). These findings align with previous results, further supporting that TBT-CpG NaVs could enhance antigen presentation through the MHC II pathway, thereby promoting DCs maturation to initiate an immune response. KEGG and Reactome analyses were also conducted to investigate the signaling pathways modulated by TBT-CpG NaVs in BMDCs. KEGG enrichment analysis showed that upregulated genes were predominantly associated with the regulation of cytokine-cytokine receptor interactions, NF-kappa B signaling pathway. Key genes such as CD40, LTa, TNF, CXCL1, CXCL2, and CXCL2 were involved in both signaling pathways, underscoring their importance in inducing an immune response (Fig. [110]4C). Reactome analysis highlighted that these differential genes were primarily involved in the regulation of the immune system (Fig. [111]4D). Overall, these results suggested that TBT-CpG NaVs induce significantly changes in the cellular mRNA transcriptome, playing a crucial role in modulating the immune system and immune response. TBT-CpG NaVs induce specific immune responses and immunoprotection To evaluate the immune response induced by TBT-CpG NaVs, mice were vaccinated with a dose of 30 µg per injection. Control groups included PBS, free TBT, and TBT + CpG, and immune responses along with protective efficacy were assessed after three immunizations (Fig. [112]5A). Results showed that the specific antibody titer induced by the nanoparticle vaccine TBT-CpG NaVs was significantly higher than that of the other groups on day 35 (Fig. [113]5B) and remained elevated on day 56 (Fig. [114]S6). ELISPOT analysis of IFN-γ (Fig. [115]5C) and IL-4 (Fig. [116]5D) secretion revealed a significantly stronger cellular immune response in the TBT-CpG NaVs group compared to PBS, TBT, and TBT + CpG groups. Fig. 5. [117]Fig. 5 [118]Open in a new tab TBT-CpG NaVs induce specific immune responses. A Schematic diagram illustrating the immunization procedure for nanovaccine administration in BALB/c mice and suckling mice. B Measurement of serum-specific IgG antibody titers on day 35 post-initial immunization, using a serial dilution method. C-D ELISPOT assay to detect the number of IFN-γ (C) and IL-4 (D) spot-forming cells following 48-hour in vitro stimulation with peptide TBT, including statistical analysis. Images were obtained and quantified using an ELISPOT reader. E-F Survival rates of 3-day-old BALB/c suckling mice infected with DENV-2 or DENV-4 (E) and 1-day-old newborn mice infected with ZIKV (F) over a 15-day observation period, with survival rate expressed as the percentage of survivors. Data are shown as mean ± SD (n = 3). Statistical analysis was performed using one-way ANOVA. **p < 0.01; ***p < 0.001 To assess protective efficacy, serum from immunized mice was mixed with DENV-2 or DENV-4 and used to infect 3-day-old Balb/c suckling mouse. Similarly, serum mixed with ZIKV was used to infect 1-day-old C57BL/6J newborn mice, with survival monitored over 15 days. The survival rate of the TBT-CpG NaVs-immunized group was significantly higher than that of the control group when infected with DENV (Fig. [119]5E) or ZIKV (Fig. [120]5F). These findings suggested that, unlike the free TBT peptide vaccine, TBT-CpG NaVs offer enhanced protective efficacy against DENV and ZIKV infections, demonstrating significant antiviral potential. Safety assessment of TBT-CpG NaVs Ensuring biosecurity is essential for clinical applications. To assess the safety of the self-assembled nanovaccine, both in vitro and in vivo toxicity evaluations were conducted. The CCK-8 assay indicated a slight dose-dependent effect, with cell viability remaining above 85% even at a TBT concentration of 600 nM, suggesting that TBT-CpG NaVs exhibit no significant cytotoxicity to BMDCs and demonstrate good safety and biocompatibility (Fig. [121]6A). In immunized mice, the biochemical indicators alanine aminotransferase (ALT) and aspartate aminotransferase (AST) remained within normal ranges (Fig. [122]6B). Furthermore, histological analysis revealed no significant damage or inflammatory response in major organs (heart, liver, spleen, lung, kidney) in mice treated with the nanovaccine (Fig. [123]6C). These results indicated that our self-assembled TBT-CpG NaVs possess a high level of safety and potential suitability for clinical research. Fig. 6. [124]Fig. 6 [125]Open in a new tab Safety evaluation of TBT-CpG NaVs. A Cell viability of BMDCs incubated with nanoparticles at various concentrations for 24 h. B Biochemical analysis of AST and ALT levels on day 7 following the final immunization. C Hematoxylin and Eosin (H&E) staining to assess histological changes in the main organs of mice on day 7 post-final immunization. Data are presented as mean ± SD (n = 3). Statistical analysis was performed using one-way ANOVA. ns, p > 0.05 Discussion Flaviviruses are becoming more prevalent globally due to uncontrolled urbanization and globalization, leading to a rise in cases and presenting new challenges for disease prevention and control [[126]36–[127]38]. This underscores the urgent need for a safe and effective vaccine to curb the spread of flaviviruses. In recent years, self-assembled nanoparticle platforms have garnered attention due to their biocompatibility, biodegradability, ease of production [[128]39], and ability to be effectively recognized by antigen-presenting cells (APCs) [[129]15]. These platforms have been widely used for anti-tumor and antiviral therapy, positioning them as a promising direction for next-generation subunit vaccine development [[130]35, [131]40, [132]41]. The use of self-assembled nanoparticle platforms combining adjuvants with epitope peptide antigens specifically for flaviviruses remains underexplored. In this study, we developed a multi-epitope peptide nanovaccine delivery system conjugated with the adjuvant CpG ODN. Our findings demonstrate that CpG ODN, serving as an adjuvant, can be covalently linked to an immunodominant epitope peptide to form a positively charged spherical nanovaccine. This spherical nanovaccine mimics the structure of flaviviruses, facilitating uptake by BMDCs. The electrostatic attraction between the positively charged nanoparticles, which have high antigen density, and the negatively charged cell surfaces promotes effective binding and internalization of the antigen [[133]42, [134]43]. Efficient delivery of peptide epitope antigens is essential to boost vaccine immunogenicity. Once internalized, these peptide epitopes stimulate the maturation of DCs and enable antigen presentation to CD4^+ and CD8^+ T cells via MHC molecules, inducing a highly targeted immune response [[135]34, [136]44]. Extensive research has also shown that nanovaccines can deliver antigens to specific tissues, improving the bioavailability of antigens, activating the body’s humoral and cellular immunity, and inducing a robust immune response [[137]45, [138]46]. Notably, our bioinformatics analysis revealed that BMDCs presented epitope peptides via the MHC II pathway and identified that TBT-CpG NaVs regulated pathways related to BMDC activation and T cell proliferation, which are crucial for mounting an effective immune response. The self-assembled nanovaccine TBT-CpG NaVs effectively activated the immune system in mice, significantly elevated serum IgG levels, and stimulated the spleen to secrete IFN-γ and IL-4, thereby enhancing antiviral capability. Epitope-based subunit vaccines are known for their specificity, safety, and reproducibility [[139]47]. Selecting appropriate antigenic epitopes is essential for optimizing the immunogenicity of these vaccines, with conserved regions within viral protein sequences serving as primary targets [[140]15]. B cell epitopes bind to B cell receptors, triggering the humoral immune response and producing specific antibodies that help resist viral invasion [[141]48]. In flaviviruses, the envelope (E) protein is the main target for neutralizing antibodies, providing long-term immunity and protection following natural infection or vaccination [[142]49]. Thus, selecting B cell epitopes derived from the E protein of flaviviruses can enhance humoral immune activation. The B cell epitope VDRGWGNGCGLFGKG, conserved across DENV1-4, ZIKV, and other flaviviruses, effectively produces neutralizing antibodies and induces broad-spectrum immunity [[143]5, [144]50]. Our neutralization test results indicated that immune serum neutralized both ZIKV and DENV, improving the survival rate of suckling mice and enhancing resistance to viral infection. The selected B cell epitopes demonstrated efficacy in producing neutralizing antibodies with broad-spectrum immune effects. It is worth noting that to further enhance the immunogenicity and stability of the vaccine, we added T-helper epitopes on both sides of the B-cell epitope with flexible peptides glycine G and leucine L. The addition of T1 helper epitope (KYVKQNTLKLAT) and T2 helper epitope (LEYIPEITLPVIAALSIAES) not only extended the length of the peptide to reduce the risk of degradation but also banded with MHC II molecules. This configuration facilitated antibody-mediated humoral immunity and effectively activated cellular immunity [[145]51]. An ideal vaccine should induce a robust immune response while maintaining safety and non-toxicity [[146]45]. Self-assembled nanovaccines, which do not rely on external carriers, reduced potential systemic toxicity and simplified preparation, opening up a new field for vaccine delivery systems [[147]30]. In our study, both in vitro and in vivo safety assessments showed no significant cytotoxic effects or inflammatory responses, confirming the favorable safety profile of TBT-CpG NaVs—an essential factor for clinical application. This nano-delivery system proved to be stable and effectively activated both cellular and humoral immune responses to exert antiviral effects. Our nanovaccine demonstrated potent immune responses against Dengue and Zika viruses; however, further studies are necessary to evaluate its efficacy against other flaviviruses. In conclusion, this study highlighted the potential of TBT-CpG NaVs in inducing balanced humoral and cellular immune responses, offering a promising strategy for flavivirus vaccine development. Future research should focus on optimizing the vaccine formulation, evaluating its efficacy across a broader range of flaviviruses, and conducting clinical trials to establish its safety and effectiveness in humans. Conclusion In summary, our study showed that the self-assembled nanovaccine TBT-CpG NaVs, possessed excellent biocompatibility and a favorable safety profile. TBT-CpG NaVs effectively stimulated the maturation of dendritic cells, promoting antigen uptake and presentation, and induced robust immune responses with high antibody titers and strong cellular immunity. These responses included the production of effective neutralizing antibodies, conferring a broad-spectrum antiviral effect against flaviviruses. Our results indicate that this self-assembled nanovaccine offers significant advantages in immunogenicity and safety, holding promising potential for combating flavivirus infections. Electronic supplementary material Below is the link to the electronic supplementary material. [148]Supplementary Material 1^ (1.8MB, docx) Acknowledgements